Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunother ; 45(8): 335-348, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35791438

ABSTRACT

Lucitanib is a multi-tyrosine kinase inhibitor whose targets are associated with angiogenesis and other key cancer and immune pathways. Its antiangiogenic properties are understood, but lucitanib's immunomodulatory activity is heretofore unknown. Lucitanib exhibited such activity in vivo, increasing CD3 + , CD8 + , and CD4 + T cells and decreasing dendritic cells and monocyte-derived suppressor cells in mouse spleens. Depletion of CD8 + T cells from syngeneic MC38 colon tumor-bearing mice reduced the antitumor efficacy of lucitanib and revealed a CD8 + T-cell-dependent component of lucitanib's activity. The combination of lucitanib and costimulatory immune pathway agonists targeting 4-1BB, glucocorticoid-induced TNFR (GITR), inducible T-cell co-stimulator (ICOS), or OX40 exhibited enhanced antitumor activity compared with each single agent in immunocompetent tumor models. Lucitanib combined with blockade of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or programmed cell death protein-1 (PD-1) coinhibitory immune pathways also showed enhanced antitumor activity over the single agents in multiple models. In CT26 tumors, lucitanib, alone or combined with anti-PD-1, reduced CD31 + vessels and depleted F4/80 + macrophages. Combination treatment also increased the number of intratumoral T cells. Gene expression in pathways associated with immune activity was upregulated by lucitanib in MC38 tumors and further potentiated by combination with anti-PD-1. Accordingly, lucitanib, alone or combined with anti-PD-1, increased intratumoral CD8 + T-cell abundance. Lucitanib's antitumor and pharmacodynamic activity, alone or combined with anti-PD-1, was not recapitulated by specific vascular endothelial growth factor receptor-2 (VEGFR2) inhibition. These data indicate that lucitanib can modulate vascular and immune components of the tumor microenvironment and cooperate with immunotherapy to enhance antitumor efficacy. They support the clinical development of lucitanib combined with immune pathway modulators to treat cancer.


Subject(s)
Antineoplastic Agents , Neoplasms , Quinolines , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Immunologic Factors/therapeutic use , Mice , Naphthalenes , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Tumor Microenvironment
2.
Haematologica ; 105(11): 2584-2591, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33131247

ABSTRACT

Antibody drug conjugates represent an important class of anti-cancer drugs in both solid tumors and hematological cancers. Here, we report preclinical data on the anti-tumor activity of the first-in-class antibody drug conjugate MEN1309/OBT076 targeting CD205. The study included preclinical in vitro activity screening on a large panel of cell lines, both as single agent and in combination and validation experiments on in vivo models. CD205 was first shown frequently expressed in lymphomas, leukemias and multiple myeloma by immunohistochemistry on tissue microarrays. Anti-tumor activity of MEN1309/OBT076 as single agent was then shown across 42 B-cell lymphoma cell lines with a median IC50 of 200 pM and induction of apoptosis in 25/42 (59.5%) of the cases. The activity appeared highly correlated with its target expression. After in vivo validation as the single agent, the antibody drug conjugate synergized with the BCL2 inhibitor venetoclax, and the anti-CD20 monoclonal antibody rituximab. The first-in-class antibody drug targeting CD205, MEN1309/OBT076, demonstrated strong pre-clinical anti-tumor activity in lymphoma, warranting further investigations as a single agent and in combination.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Lymphoma , Antibodies, Monoclonal/pharmacology , Antigens, CD20 , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Lymphoma/drug therapy , Rituximab/therapeutic use
3.
Mol Cancer Ther ; 18(9): 1533-1543, 2019 09.
Article in English | MEDLINE | ID: mdl-31227646

ABSTRACT

CD205 is a type I transmembrane glycoprotein and is a member of the C-type lectin receptor family. Analysis by mass spectrometry revealed that CD205 was robustly expressed and highly prevalent in a variety of solid malignancies from different histotypes. IHC confirmed the increased expression of CD205 in pancreatic, bladder, and triple-negative breast cancer (TNBC) compared with that in the corresponding normal tissues. Using immunofluorescence microscopy, rapid internalization of the CD205 antigen was observed. These results supported the development of MEN1309/OBT076, a fully humanized CD205-targeting mAb conjugated to DM4, a potent maytansinoid derivate, via a cleavable N-succinimidyl-4-(2-pyridyldithio) butanoate linker. MEN1309/OBT076 was characterized in vitro for target binding affinity, mechanism of action, and cytotoxic activity against a panel of cancer cell lines. MEN1309/OBT076 displayed selective and potent cytotoxic effects against tumor cells exhibiting strong and low to moderate CD205 expression. In vivo, MEN1309/OBT076 showed potent antitumor activity resulting in durable responses and complete tumor regressions in many TNBC, pancreatic, and bladder cancer cell line-derived and patient-derived xenograft models, independent of antigen expression levels. Finally, the pharmacokinetics and pharmacodynamic profile of MEN1309/OBT076 was characterized in pancreatic tumor-bearing mice, demonstrating that the serum level of antibody-drug conjugate (ADC) achieved through dosing was consistent with the kinetics of its antitumor activity. Overall, our data demonstrate that MEN1309/OBT076 is a novel and selective ADC with potent activity against CD205-positive tumors. These data supported the clinical development of MEN1309/OBT076, and further evaluation of this ADC is currently ongoing in the first-in-human SHUTTLE clinical trial.


Subject(s)
Immunoconjugates/pharmacology , Lectins, C-Type/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Receptors, Cell Surface/antagonists & inhibitors , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, CD/metabolism , CHO Cells , Cell Line, Tumor , Cricetulus , Female , HEK293 Cells , HT29 Cells , Humans , Immunoconjugates/chemistry , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , MCF-7 Cells , Maytansine/chemistry , Maytansine/pharmacology , Mice , Mice, Nude , Mice, SCID , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism
4.
Genes Dev ; 27(9): 1016-31, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651856

ABSTRACT

The mechanisms by which the p53 tumor suppressor acts remain incompletely understood. To gain new insights into p53 biology, we used high-throughput sequencing to analyze global p53 transcriptional networks in primary mouse embryo fibroblasts in response to DNA damage. Chromatin immunoprecipitation sequencing reveals 4785 p53-bound sites in the genome located near 3193 genes involved in diverse biological processes. RNA sequencing analysis shows that only a subset of p53-bound genes is transcriptionally regulated, yielding a list of 432 p53-bound and regulated genes. Interestingly, we identify a host of autophagy genes as direct p53 target genes. While the autophagy program is regulated predominantly by p53, the p53 family members p63 and p73 contribute to activation of this autophagy gene network. Induction of autophagy genes in response to p53 activation is associated with enhanced autophagy in diverse settings and depends on p53 transcriptional activity. While p53-induced autophagy does not affect cell cycle arrest in response to DNA damage, it is important for both robust p53-dependent apoptosis triggered by DNA damage and transformation suppression by p53. Together, our data highlight an intimate connection between p53 and autophagy through a vast transcriptional network and indicate that autophagy contributes to p53-dependent apoptosis and cancer suppression.


Subject(s)
Autophagy/genetics , DNA Damage/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Animals , Cell Cycle Checkpoints/genetics , Cell Survival/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Genome-Wide Association Study , Mice , Protein Binding , Sequence Analysis, RNA
5.
Breast Cancer Res ; 14(2): R65, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22515648

ABSTRACT

INTRODUCTION: Perp is a transcriptional target of both p53 during DNA damage-induced apoptosis and p63 during stratified epithelial development. Perp-/- mice exhibit postnatal lethality associated with dramatic blistering of the epidermis and oral mucosa, reflecting a critical role in desmosome-mediated intercellular adhesion in keratinocytes. However, the role of Perp in tissue homeostasis in other p63-dependent stratified epithelial tissues is poorly understood. Given that p63 is essential for proper mammary gland development and that cell adhesion is fundamental for ensuring the proper architecture and function of the mammary epithelium, here we investigate Perp function in the mammary gland. METHODS: Immunofluorescence and Western blot analysis were performed to characterize Perp expression and localization in the mouse mammary epithelium throughout development. The consequences of Perp deficiency for mammary epithelial development and homeostasis were examined by using in vivo mammary transplant assays. Perp protein levels in a variety of human breast cancer cell lines were compared with those in untransformed cells with Western blot analysis. The role of Perp in mouse mammary tumorigenesis was investigated by aging cohorts of K14-Cre/+;p53fl/fl mice that were wild-type or deficient for Perp. Mammary tumor latency was analyzed, and tumor-free survival was assessed using Kaplan-Meier analysis. RESULTS: We show that Perp protein is expressed in the mammary epithelium, where it colocalizes with desmosomes. Interestingly, although altering desmosomes through genetic inactivation of Perp does not dramatically impair mammary gland ductal development, Perp loss affects mammary epithelial homeostasis by causing the accumulation of inflammatory cells around mature mammary epithelium. Moreover, we show reduced Perp expression in many human breast cancer cell lines compared with untransformed cells. Importantly, Perp deficiency also promotes the development of mouse mammary cancer. CONCLUSIONS: Together, these observations demonstrate an important role for Perp in normal mammary tissue function and in mammary cancer suppression. In addition, our findings highlight the importance of desmosomes in cancer suppression and suggest the merit of evaluating Perp as a potential prognostic indicator or molecular target in breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Genes, p53 , Mammary Glands, Animal/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Desmosomes/metabolism , Epithelial Cells/metabolism , Female , Genes, Tumor Suppressor , Homeostasis , Keratin-14/genetics , Mammary Glands, Animal/cytology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/genetics , Trans-Activators/genetics
6.
Nat Rev Cancer ; 11(5): 317-23, 2011 May.
Article in English | MEDLINE | ID: mdl-21508970

ABSTRACT

Adherens junctions, which are intercellular adhesive complexes that are crucial for maintaining epithelial homeostasis, are downregulated in many cancers to promote tumour progression. However, the role of desmosomes - adhesion complexes that are related to adherens junctions - in carcinogenesis has remained elusive. Recent studies using mouse genetic approaches have uncovered a role for desmosomes in tumour suppression, demonstrating that desmosome downregulation occurs before that of adherens junctions to drive tumour development and early invasion, suggesting a two-step model of adhesion dysfunction in cancer progression.


Subject(s)
Desmosomes/physiology , Neoplastic Processes , Tumor Suppressor Proteins/physiology , Adherens Junctions/physiology , Animals , Humans
7.
PLoS Genet ; 6(10): e1001168, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20975948

ABSTRACT

Dysregulated cell-cell adhesion plays a critical role in epithelial cancer development. Studies of human and mouse cancers have indicated that loss of adhesion complexes known as adherens junctions contributes to tumor progression and metastasis. In contrast, little is known regarding the role of the related cell-cell adhesion junction, the desmosome, during cancer development. Studies analyzing expression of desmosome components during human cancer progression have yielded conflicting results, and therefore genetic studies using knockout mice to examine the functional consequence of desmosome inactivation for tumorigenesis are essential for elucidating the role of desmosomes in cancer development. Here, we investigate the consequences of desmosome loss for carcinogenesis by analyzing conditional knockout mice lacking Perp, a p53/p63 regulated gene that encodes an important component of desmosomes. Analysis of Perp-deficient mice in a UVB-induced squamous cell skin carcinoma model reveals that Perp ablation promotes both tumor initiation and progression. Tumor development is associated with inactivation of both of Perp's known functions, in apoptosis and cell-cell adhesion. Interestingly, Perp-deficient tumors exhibit widespread downregulation of desmosomal constituents while adherens junctions remain intact, suggesting that desmosome loss is a specific event important for tumorigenesis rather than a reflection of a general change in differentiation status. Similarly, human squamous cell carcinomas display loss of PERP expression with retention of adherens junctions components, indicating that this is a relevant stage of human cancer development. Using gene expression profiling, we show further that Perp loss induces a set of inflammation-related genes that could stimulate tumorigenesis. Together, these studies suggest that Perp-deficiency promotes cancer by enhancing cell survival, desmosome loss, and inflammation, and they highlight a fundamental role for Perp and desmosomes in tumor suppression. An understanding of the factors affecting cancer progression is important for ultimately improving the diagnosis, prognostication, and treatment of cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Desmosomes/metabolism , Membrane Proteins/genetics , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tissue Array Analysis , Trans-Activators/genetics , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
8.
J Cell Biol ; 185(7): 1243-58, 2009 Jun 29.
Article in English | MEDLINE | ID: mdl-19546243

ABSTRACT

Dsg1 (desmoglein 1) is a member of the cadherin family of Ca(2+)-dependent cell adhesion molecules that is first expressed in the epidermis as keratinocytes transit out of the basal layer and becomes concentrated in the uppermost cell layers of this stratified epithelium. In this study, we show that Dsg1 is not only required for maintaining epidermal tissue integrity in the superficial layers but also supports keratinocyte differentiation and suprabasal morphogenesis. Dsg1 lacking N-terminal ectodomain residues required for adhesion remained capable of promoting keratinocyte differentiation. Moreover, this capability did not depend on cytodomain interactions with the armadillo protein plakoglobin or coexpression of its companion suprabasal cadherin, Dsc1 (desmocollin 1). Instead, Dsg1 was required for suppression of epidermal growth factor receptor-Erk1/2 (extracellular signal-regulated kinase 1/2) signaling, thereby facilitating keratinocyte progression through a terminal differentiation program. In addition to serving as a rigid anchor between adjacent cells, this study implicates desmosomal cadherins as key components of a signaling axis governing epithelial morphogenesis.


Subject(s)
Cell Differentiation/physiology , Desmoglein 1/metabolism , Epidermis/physiology , ErbB Receptors/metabolism , Keratinocytes/physiology , Morphogenesis/physiology , Signal Transduction/physiology , Cell Adhesion/physiology , Cells, Cultured , Desmoglein 1/genetics , Enzyme Activation , Epidermal Cells , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Keratinocytes/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tissue Culture Techniques
9.
J Invest Dermatol ; 129(7): 1710-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19158843

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune bullous disease in which autoantibodies against proteins of the desmosomal adhesion complex perturb desmosomal function, leading to intercellular adhesion defects in the oral mucosa and skin. Previous studies have demonstrated a central role for downregulation of the desmosomal cadherin desmoglein 3 (DSG3) in the pathogenesis of PV. However, the effects of non-cadherin desmosomal proteins in modulating the cellular manifestations of PV remain poorly understood. Here, we characterize the expression and functional importance of Perp, a newly discovered tetraspan desmosomal protein, in PV. Our data demonstrate that PV autoantibodies disrupt Perp expression at the membrane and trigger its internalization along with DSG3 into the endosomal pathway, where it is ultimately targeted to the lysosome for degradation. We further show that Perp deficiency exacerbates the pathogenic effects of PV autoantibodies on keratinocytes by enhancing both the depletion of desmosomal DSG3 and intercellular adhesion defects. Together, our findings highlight the importance of non-cadherin desmosomal proteins in modulating PV phenotypes and provide new insight into Perp's role in the desmosome.


Subject(s)
Autoantibodies/immunology , Desmosomes/immunology , Keratinocytes/immunology , Membrane Proteins/immunology , Pemphigus/immunology , Animals , Cell Adhesion/immunology , Cells, Cultured , Desmoglein 3/immunology , Desmoglein 3/metabolism , Desmosomes/metabolism , Endosomes/immunology , Genes, Tumor Suppressor , Humans , Immunoglobulin G/immunology , Keratinocytes/cytology , Keratinocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Pemphigus/metabolism , Pemphigus/physiopathology , Phenotype
10.
J Dermatol Sci ; 45(1): 7-21, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17141479

ABSTRACT

The desmosomal cadherins, which include desmogleins and desmocollins, are Ca(2+)-dependent adhesion molecules that cooperate to make up the adhesive core of intercellular junctions known as desmosomes. The roles of desmosomal cadherins in epidermal integrity and as targets in human cutaneous disease have been well established. However, the molecular basis of these disorders is still poorly understood, due in part to a lack of fundamental knowledge about the organization of the adhesive interface and molecular machinery that dictates the proper presentation of desmogleins and desmocollins on the cell surface. Further, the diversity of the desmosomal cadherin family, and their individualized expression patterns within complex tissues, suggests that these adhesion molecules may have differentiation-specific functions that transcend their roles in intercellular adhesion. Here we will review the most recent data from our own group and others that are beginning to unveil the diverse properties and functions of this complex family of adhesion molecules.


Subject(s)
Desmosomal Cadherins/physiology , Animals , Cell Adhesion/physiology , Desmosomal Cadherins/chemistry , Desmosomal Cadherins/genetics , Desmosomes/physiology , Embryo, Mammalian/physiology , Epidermis/growth & development , Humans , Molecular Structure
11.
J Invest Dermatol ; 127(4): 792-801, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17110936

ABSTRACT

The armadillo family protein plakoglobin (Pg) is a well-characterized component of anchoring junctions, where it functions to mediate cell-cell adhesion and maintain epithelial tissue integrity. Although its closest homolog beta-catenin acts in the Wnt signaling pathway to dictate cell fate and promote proliferation and survival, the role of Pg in these processes is not well understood. Here, we investigate how Pg affects the survival of mouse keratinocytes by challenging both Pg-null cells and their heterozygote counterparts with apoptotic stimuli. Our results indicate that Pg deletion protects keratinocytes from apoptosis, with null cells exhibiting delayed mitochondrial cytochrome c release and activation of caspase-3. Pg-null keratinocytes also exhibit increased messenger RNA and protein levels of the anti-apoptotic molecule Bcl-X(L) compared to heterozygote controls. Importantly, reintroduction of Pg into the null cells shifts their phenotype towards that of the Pg+/- keratinocytes, providing further evidence that Pg plays a direct role in regulating cell survival. Taken together, our results suggest that in addition to its adhesive role in epithelia, Pg may also function in contrast to the pro-survival tendencies of beta-catenin, to potentiate death in cells damaged by apoptotic stimuli, perhaps limiting the potential for the propagation of mutations and cellular transformation.


Subject(s)
Apoptosis/physiology , Keratinocytes/physiology , gamma Catenin/physiology , Animals , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation/physiology , Keratinocytes/cytology , Mice , Mice, Knockout , Mitochondria/metabolism , RNA, Messenger/metabolism , Time Factors , bcl-X Protein/genetics , bcl-X Protein/metabolism , gamma Catenin/deficiency
12.
J Biol Chem ; 281(6): 3614-24, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16286477

ABSTRACT

Although a number of cell adhesion proteins have been identified as caspase substrates, the potential role of differentiation-specific desmosomal cadherins during apoptosis has not been examined. Here, we demonstrate that UV-induced caspase cleavage of the human desmoglein 1 cytoplasmic tail results in distinct 17- and 140- kDa products, whereas metalloproteinase-dependent shedding of the extracellular adhesion domain generates a 75-kDa product. In vitro studies identify caspase-3 as the preferred enzyme that cleaves desmoglein 1 within its unique repeating unit domain at aspartic acid 888, part of a consensus sequence not conserved among the other desmosomal cadherins. Apoptotic processing leads to decreased cell surface expression of desmoglein 1 and re-localization of its C terminus diffusely throughout the cytoplasm over a time course comparable with the processing of other desmosomal proteins and cytoplasmic keratins. Importantly, whereas classic cadherins have been reported to promote cell survival, short hairpin RNA-mediated suppression of desmoglein 1 in differentiated keratinocytes protected cells from UV-induced apoptosis. Collectively, our results identify desmoglein 1 as a novel caspase and metalloproteinase substrate whose cleavage likely contributes to the dismantling of desmosomes during keratinocyte apoptosis and also reveal desmoglein 1 as a previously unrecognized regulator of apoptosis in keratinocytes.


Subject(s)
Apoptosis , Caspases/metabolism , Desmoglein 1/physiology , Gene Expression Regulation, Enzymologic , Keratinocytes/enzymology , Binding Sites , Blotting, Western , Caspase 3 , Cell Differentiation , Cell Line, Tumor , Cytoplasm/metabolism , DNA, Complementary/metabolism , Desmoglein 1/metabolism , Desmosomes/metabolism , Doxycycline/pharmacology , Humans , Indoles/pharmacology , Keratinocytes/metabolism , Microscopy, Fluorescence , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA/chemistry , Retroviridae/genetics , Time Factors , Transfection , Ultraviolet Rays
13.
Differentiation ; 72(8): 419-33, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15606501

ABSTRACT

Desmoglein 1 (Dsg1) is a component of desmosomes present in the upper epidermis and can be targeted by autoimmune antibodies or bacterial toxins, resulting in skin blistering diseases. These defects in tissue integrity are believed to result from compromised desmosomal adhesion; yet, previous attempts to directly test the adhesive roles of desmosomal cadherins using normally non-adherent L cells have yielded mixed results. Here, two complementary approaches were used to better resolve the molecular determinants for Dsg1-mediated adhesion: (1) a tetracycline-inducible system was used to modulate the levels of Dsg1 expressed in L cell lines containing desmocollin 1 (Dsc1) and plakoglobin (PG) and (2) a retroviral gene delivery system was used to introduce Dsg1 into normal human epidermal keratinocytes (NHEK). By increasing Dsg1 expression relative to Dsc1 and PG, we were able to demonstrate that the ratio of Dsg1:Dsc1 is a critical determinant of desmosomal adhesion in fibroblasts. The distribution of Dsg1 was organized at areas of cell-cell contact in the multicellular aggregates that formed in these suspension cultures. Similarly, the introduction of Dsg1 into NHEKs was capable of increasing the aggregation of single cell suspensions and further enhanced the adhesive strength of intact epithelial sheets. Endogenous Dsc1 levels were also increased in NHEKs containing Dsg1, providing further support for the coordination of these two desmosomal cadherins in regulating adhesive structures. These Dsg1-mediated effects on intercellular adhesion were directly related to the presence of an intact extracellular domain as ETA, a toxin that specifically cleaves this desmosomal cadherin, inhibited adhesion in both fibroblasts and keratinocytes. Collectively, these observations demonstrate that Dsg1 promotes the formation of intercellular adhesion complexes and suggest that the relative level of Dsg and Dsc expressed at the cell surface regulates this adhesive process.


Subject(s)
Cadherins/metabolism , Cell Adhesion/physiology , Desmosomes/physiology , Membrane Glycoproteins/metabolism , Animals , Cadherins/analysis , Cadherins/genetics , Cytoskeletal Proteins/metabolism , Desmocollins , Desmoglein 1 , Desmogleins , Desmoplakins , Fibroblasts/chemistry , Fibroblasts/drug effects , Humans , Keratinocytes/metabolism , L Cells , Membrane Glycoproteins/genetics , Mice , Tetracycline/pharmacology , Up-Regulation , gamma Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...