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










Publication year range
1.
Front Genet ; 7: 171, 2016.
Article in English | MEDLINE | ID: mdl-27757122

ABSTRACT

Quiescence is the prevailing state of many cell types under homeostatic conditions. Yet, surprisingly little is known about how quiescent cells respond to energetic and metabolic challenges. To better understand compensatory responses of quiescent cells to metabolic stress, we established, in human primary dermal fibroblasts, an experimental 'energy restriction' model. Quiescence was achieved by short-term culture in serum-deprived media and ATP supply restricted using a combination of glucose transport inhibitors and mitochondrial uncouplers. In aggregate, these measures led to markedly reduced intracellular ATP levels while not compromising cell viability over the observation period of 48 h. Analysis of the transcription factor (TF) landscape induced by this treatment revealed alterations in several signal transduction nodes beyond the expected biosynthetic adaptations. These included increased abundance of NF-κB regulated TFs and altered TF subsets regulated by Akt and p53. The observed changes in gene regulation and corresponding alterations in key signaling nodes are likely to contribute to cell survival at intracellular ATP concentrations substantially below those achieved by growth factor deprivation alone. This experimental model provides a benchmark for the investigation of cell survival pathways and related molecular targets that are associated with restricted energy supply associated with biological aging and metabolic diseases.

2.
BMC Dev Biol ; 10: 102, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20929554

ABSTRACT

BACKGROUND: The p53 protein family coordinates stress responses of cells and organisms. Alternative promoter usage and/or splicing of p53 mRNA gives rise to at least nine mammalian p53 proteins with distinct N- and C-termini which are differentially expressed in normal and malignant cells. The human N-terminal p53 variants contain either the full-length (FL), or a truncated (ΔN/Δ40) or no transactivation domain (Δ133) altogether. The functional consequences of coexpression of the different p53 isoforms are poorly defined. Here we investigated functional aspects of the zebrafish ΔNp53 ortholog in the context of FLp53 and the zebrafish Δ133p53 ortholog (Δ113p53) coexpressed in the developing embryo. RESULTS: We cloned the zebrafish ΔNp53 isoform and determined that ionizing radiation increased expression of steady-state ΔNp53 and Δ113p53 mRNA levels in zebrafish embryos. Ectopic ΔNp53 expression by mRNA injection caused hypoplasia and malformation of the head, eyes and somites, yet partially counteracted lethal effects caused by concomitant expression of FLp53. FLp53 expression was required for developmental aberrations caused by ΔNp53 and for ΔNp53-dependent expression of the cyclin-dependent kinase inhibitor 1A (CDKN1A, p21, Cip1, WAF1). Knockdown of p21 expression markedly reduced the severity of developmental malformations associated with ΔNp53 overexpression. By contrast, forced Δ113p53 expression had little effect on ΔNp53-dependent embryonal phenotypes. These functional attributes were shared between zebrafish and human ΔNp53 orthologs ectopically expressed in zebrafish embryos. All 3 zebrafish isoforms could be coimmunoprecipitated with each other after transfection into Saos2 cells. CONCLUSIONS: Both alternative N-terminal p53 isoforms were expressed in developing zebrafish in response to cell stress and antagonized lethal effects of FLp53 to different degrees. However, in contrast to Δ113p53, forced ΔNp53 expression itself led to developmental defects which depended, in part, on p21 transactivation. In contrast to FLp53, the developmental abnormalities caused by ΔNp53 were not counteracted by concomitant expression of Δ113p53.


Subject(s)
Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Base Sequence , Cell Line , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Humans , Molecular Sequence Data , Protein Isoforms/genetics , RNA Caps , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation, Ionizing , Tumor Suppressor Protein p53/genetics , Zebrafish/anatomy & histology , Zebrafish/genetics
3.
Cancer Biol Ther ; 8(22): 2147-52, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19783899

ABSTRACT

Therapeutic antibodies frequently cause side effects by binding antigen in non-target tissues. Here we demonstrate a novel molecular design of antibodies that addresses this problem by reversibly "masking" antibody complementarity determining regions until they reach diseased tissues containing disease-associated proteases. Specifically, two distinct single-chain Fv (scFv) fragments derived from antibodies against the epidermal growth factor receptor (cetuximab and 425) were fused a protease susceptible linker to their epitopes, which were engineered to encourage intermolecular association. Surface plasmon resonance and flow cytometry were used to confirm that the masked complex poorly interacts with native antigen, whereas protease treatment restores antigen recognition. Minimally, the "masked" scFvs possesses an eight-fold lower association with the epitope compared with the individual scFvs unmasked by proteolytic cleavage. This molecular design may have general utility for targeted release of therapeutic antibodies at disease sites.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibody Affinity , Cross-Linking Reagents/chemistry , Drug Design , ErbB Receptors/immunology , Matrix Metalloproteinase 9/metabolism , Oligopeptides/chemistry , Prodrugs/chemistry , Recombinant Fusion Proteins/chemistry , Single-Chain Antibodies/chemistry , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Complementarity Determining Regions/chemistry , Cross-Linking Reagents/metabolism , Dimerization , Drug Delivery Systems , Drug Synergism , Epitopes/genetics , Epitopes/immunology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Hydrolysis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Oligopeptides/metabolism , Point Mutation , Prodrugs/administration & dosage , Protein Structure, Tertiary , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , Substrate Specificity , Surface Plasmon Resonance
4.
Immun Ageing ; 5: 5, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18631391

ABSTRACT

BACKGROUND: Chronic inflammation is a well-known corollary of the aging process and is believed to significantly contribute to morbidity and mortality of many age-associated chronic diseases. However, the mechanisms that cause age-associated inflammatory changes are not well understood. Particularly, the contribution of cell stress responses to age-associated inflammation in 'non-inflammatory' cells remains poorly defined. The present cross-sectional study focused on differences in molecular signatures indicative of inflammatory states associated with biological aging of human fibroblasts from donors aged 22 to 92 years. RESULTS: Gene expression profiling revealed elevated steady-state transcript levels consistent with a chronic inflammatory state in fibroblast cell-strains obtained from older donors. We also observed enhanced NF-kappaB DNA binding activity in a subset of strains, and the NF-kappaB profile correlated with mRNA expression levels characteristic of inflammatory processes, which include transcripts coding for cytokines, chemokines, components of the complement cascade and MHC molecules. This intrinsic low-grade inflammatory state, as it relates to aging, occurs in cultured cells irrespective of the presence of other cell types or the in vivo context. CONCLUSION: Our results are consistent with the view that constitutive activation of inflammatory pathways is a phenomenon prevalent in aged fibroblasts. It is possibly part of a cellular survival process in response to compromised mitochondrial function. Importantly, the inflammatory gene expression signature described here is cell autonomous, i.e. occurs in the absence of prototypical immune or pro-inflammatory cells, growth factors, or other inflammatory mediators.

5.
Cancer Biol Ther ; 7(5): 726-33, 2008 May.
Article in English | MEDLINE | ID: mdl-18424917

ABSTRACT

Monoclonal antibodies (mAbs) that inhibit activation of the epidermal growth factor receptor (EGFR) have shown therapeutic potential in select malignancies including breast cancer. Here, we describe that combined use of two such mAbs, C225 (Cetuximab) and 425 (EMD55900), reduced growth and survival of EGFR overexpressing MDA-MB-468 breast cancer cells more effectively than either antibody alone. Similarly, the C225/425 antibody combination more effectively inhibited AKT and MAPK phosphorylation in MDA-MB-468 cells. Surface plasmon resonance, size exclusion chromatography and analytical ultracentrifugation demonstrated that mAbs C225 and 425 simultaneously bind to distinct antigenic epitopes on domain III of the soluble wild-type EGFR. Furthermore, neither mAb competed with the other for binding to cells expressing either wild-type EGFR or a mutant EGFR (EGFRvIII) associated with neoplasia. Mutagenesis experiments revealed that residues S460/G461 in EGFR domain III are essential components of the 425 epitope and clearly distinguish it from the EGF/ TGFalpha binding site and the C225 interaction interface. Collectively, these results support the conclusion that therapeutic EGFR blockade in cancer patients by combined use of mAbs C225 and 425 could provide advantages over the use of the two antibodies as single agents.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes , ErbB Receptors/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epitope Mapping , Epitopes/chemistry , Humans , Mice , Molecular Conformation , NIH 3T3 Cells , Tyrphostins/pharmacology
6.
Cancer Res ; 66(10): 5209-15, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707445

ABSTRACT

Previous studies addressing functional aspects of nuclear factor kappaB (NF-kappaB) activation in normal and transformed keratinocytes revealed complex and seemingly contradictory roles of this transcription factor in this cell type. In normal skin, NF-kappaB signaling seems to inhibit squamous cell carcinoma development whereas, in squamous cell carcinoma themselves, deregulated NF-kappaB expression and/or signaling is frequently observed. To further investigate this paradox, we focused on NF-kappaB activation as it relates to the transformed phenotype of immortalized but nontumorigenic human keratinocytes (HaCaT cells). We observed that NF-kappaB activity contributed to survival and growth of cultured HaCaT keratinocytes as shown by use of pharmacologic NF-kappaB inhibitors, RNA interference, and inducible overexpression of a dominant interfering IkappaB construct. NF-kappaB activation was largely provided through interaction with extracellular matrix components because preventing cell attachment by forced suspension culture markedly reduced NFkappaB signaling associated with cell death (anoikis); conversely, anoikis was partially reversed by NF-kappaB activation induced either by tumor necrosis factor-alpha treatment or by overexpressing the NF-kappaB p65 subunit in HaCaT cells. Furthermore, overexpression of NF-kappaBp65 in HaCaT cells induced colony formation in soft agar and tumorigenicity in nude mice. In summary, as opposed to normal keratinocytes, immortalized HaCaT keratinocytes provide a cellular context in which deregulated NF-kappaB signaling supports multiple malignant traits in vitro and in vivo.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Transformation, Neoplastic/metabolism , Keratinocytes/metabolism , NF-kappa B/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Adhesion/physiology , Cell Line , Cell Survival/physiology , Cell Transformation, Neoplastic/pathology , Extracellular Matrix/pathology , Humans , Keratinocytes/pathology , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , Signal Transduction , Transcription Factor RelA/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
7.
Cancer Biol Ther ; 5(5): 498-504, 2006 May.
Article in English | MEDLINE | ID: mdl-16582597

ABSTRACT

Activation of the epidermal growth factor receptor (EGFR) provides a measure of protection to immortalized epidermal keratinocytes (HaCaT cells) against apoptosis induced by diverse cellular stressors. This effect is due, in part, to sustained MAPK-dependent Bcl-xL expression. Here, we report a second EGFR/MAPK-dependent signaling event that protects HaCaT cells against apoptosis incurred during forced suspension culture (anoikis). This pathway targets Bim, a pro-apoptotic BH3-only Bcl-2 family member. Bim expression was functionally relevant to HaCaT cell survival as demonstrated by partial protection against anoikis provided by siRNA-induced Bim downregulation. Growth factor starvation of attached and suspended cells was associated with enhanced Bim expression whereas EGFR activation reduced Bim expression by inducing Bim phosphorylation and proteasomal degradation. EGFR-dependent Bim phosphorylation required MAPK activation. Furthermore, PKC-delta activity contributed to both MEK/MAPK phosphorylation and Bim phosphorylation as demonstrated using both pharmacological inhibitors of PKC-delta and siRNA-mediated PKC-delta knockdown. In addition to HaCaT cells, EGFR activation supported survival and induced Bim phosphorylation in several squamous carcinoma cell lines in a strictly MAPK-dependent fashion. These results establish that EGFR activation attenuates susceptibility of immortalized and malignant keratinocytes to apoptosis by post-translational control of Bim-EL expression through a pathway requiring PKC-delta and MEK/MAPK activation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Anoikis , Apoptosis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Down-Regulation , Humans , Keratinocytes/metabolism , MAP Kinase Kinase 1 , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Processing, Post-Translational , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology
8.
Nucleic Acids Res ; 33(15): 4914-21, 2005.
Article in English | MEDLINE | ID: mdl-16284195

ABSTRACT

During early zebrafish (Danio rerio) development zygotic transcription does not begin until the mid-blastula transition (MBT) 3 h after fertilization. MBT demarcates transition from synchronous short cell cycles of S and M phases exclusively to full cycles encompassing G1 and G2 phases. Transcriptional profiling and RT-PCR analyses during these phases enabled us to determine that this shift corresponds to decreased transcript levels of S/M phase cell cycle control genes (e.g. ccna2, ccnb1, ccnb2 and ccne) and increased transcript levels of ccnd1, encoding cyclin D1, and orthologs of p21 (p21-like) and retinoblastoma (Rb-like 1). To investigate the regulation of this process further, the translation of ccnd1 mRNA, a G1/S checkpoint control element, was impaired by microinjection of ccnd1-specific morpholino phosphorodiamidate (MO) 20mer or hydroxyprolyl-phosphono peptide nucleic acid (HypNA-pPNA) 16mer antisense oligonucleotides. The resulting downregulation of cyclin D1 protein resulted in microophthalmia and microcephaly, but not lethality. The phenotypes were not seen with 3-mismatch MO 20mers or 1-mismatch HypNA-pPNA 16mers, and were rescued by an exogenous ccnd1 mRNA construct with five mismatches. Collectively, these results indicate that transcription of key molecular determinants of asynchronous cell cycle control in zebrafish embryos commences at MBT and that the reduction of cyclin D1 expression compromises zebrafish eye and head development.


Subject(s)
Cyclin D1/genetics , Genes, cdc , Oligodeoxyribonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Zebrafish/embryology , Zebrafish/genetics , Animals , Cyclin D1/antagonists & inhibitors , Cyclin D1/physiology , Down-Regulation , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental , Morpholinos , Oligodeoxyribonucleotides/chemistry , Oligonucleotides, Antisense/chemistry , Peptide Nucleic Acids/chemistry , Transcription, Genetic , Zebrafish/metabolism
9.
Cancer Res ; 64(11): 3934-9, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15173005

ABSTRACT

Previous work implicated activation of the signal transducer and activator of transcription (STAT)3 downstream of the epidermal growth factor receptor (EGFR) in the malignant phenotype of squamous carcinoma cells (SCC). Here, we show that EGFR-dependent STAT3 activation is restricted to malignant keratinocytes. Specifically, constitutive and epidermal growth factor-induced phosphorylation of STAT3 on Y705 was observed only in SCC but not in either immortalized (HaCaT) or normal keratinocyte strains. Furthermore, STAT3 activation as determined by DNA binding assays was restricted to SCC and dependent on EGFR activation. Forced expression of EGFR in immortalized keratinocytes (HaCaT cells) was associated with enhanced EGFR activation but not STAT3-Y705 phosphorylation. EGFR-dependent activation of mitogen-activated protein kinase (MAPK) kinase 1 negatively regulated STAT3-Y705 phosphorylation in normal and malignant keratinocytes. Together, these results underscore that EGFR activation is required but not sufficient for STAT3 activation to occur in malignant keratinocytes. They also highlight complex regulation of STAT3 phosphorylation through EGFR activation including negative regulation via the MAPK kinase/MAPK signaling pathway.


Subject(s)
Carcinoma, Squamous Cell/metabolism , DNA-Binding Proteins/metabolism , Keratinocytes/metabolism , Skin Neoplasms/metabolism , Trans-Activators/metabolism , Cell Line, Tumor , DNA/metabolism , DNA, Neoplasm/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , ErbB Receptors/biosynthesis , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Humans , Keratinocytes/physiology , MAP Kinase Kinase Kinases/metabolism , Phosphorylation , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transfection
10.
Cancer Res ; 63(1): 1-5, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12517767

ABSTRACT

Aberrant activation of the epidermal growth factor receptor (EGFR) is frequently observed in neoplasia,notably in tumors of epithelial origin. Attempts to treat such tumors with EGFR antagonists have met with remarkable initial successes, particularly when EGFR antagonists were used in combination with chemotherapy or ionizing radiation. Considering the almost ubiquitous expression of the EGFR in normal epithelial tissues, these clinical trials also revealed a surprisingly low rate of adverse side effects associated with EGFR blockade. This review highlights antiapoptotic effects of EGFR activation as they relate to therapeutic efficacy of EGFR blockade. We introduce the concept that control of cell survival through EGFR activation is conditional in the sense that it is rate limiting to tumor cell survival but not to survival of normal epithelial cells. Specifically, normal epithelial cells are provided with a full complement of physiological cell-cell contacts and cell-matrix interactions that lessen their dependence on survival signals provided by the EGFR. By contrast, malignant tumor cells faced with inadequate cell-matrix contacts critically depend on EGFR activation for survival, rendering them more susceptible to apoptosis induction by EGFR blockade. Redundant control of cell survival by the EGFR and extracellular matrix/cell adhesion receptors is enabled, in part, by shared signal transduction pathways that control expression and activation states of members of the Bcl-2 family of apoptosis regulators.


Subject(s)
Apoptosis/physiology , ErbB Receptors/physiology , Neoplasms/therapy , Humans , Neoplasms/pathology , Signal Transduction
11.
Oncogene ; 21(14): 2161-70, 2002 Mar 28.
Article in English | MEDLINE | ID: mdl-11948399

ABSTRACT

The human metastasis-associated gene (MTA1), a member of the nucleosome remodeling complex with histone deacetylase activity, is frequently overexpressed in biologically aggressive epithelial neoplasms. Here, we extend this observation to squamous carcinoma cells, which express high levels of MTA1 relative to normal or immortalized keratinocytes. To address functional aspects of MTA1 expression, we established variants of human immortalized keratinocytes (HaCaT cells) by expressing MTA1 cDNA in both the sense and antisense orientations. We demonstrate that (1) forced MTA1 expression enhances migration and invasion of immortalized keratinocytes; (2) MTA1 expression is necessary but not sufficient for cell survival in the anchorage independent state; (3) MTA1 contributes to expression of the anti-apoptotic Bcl-2 family member Bcl-x(L); (4) MTA1 expression in immortalized keratinocytes depends, in part, on activation of the epidermal growth factor receptor (EGFR). These results establish that, in keratinocytes, MTA1 expression contributes to several aspects of the metastatic phenotype including survival in the anchorage independent state, migration, and invasion.


Subject(s)
Cell Movement , Histone Deacetylases , Keratinocytes/metabolism , Keratinocytes/pathology , Neoplasm Invasiveness/pathology , Proteins/metabolism , Repressor Proteins , Anoikis , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Division , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators , bcl-X Protein
12.
J Cell Sci ; 115(Pt 7): 1391-402, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11896187

ABSTRACT

P120 catenin (p120ctn) belongs to the Armadillo family of proteins, which is implicated in cell-cell adhesion and signal transduction. Owing to alternative splicing and multiple translation initiation codons, several p120ctn isoforms can be expressed from a single gene. All p120ctn isoforms share the central Armadillo repeat domain but have divergent N- and C-termini. Little is known about the biological functions of the different isoforms. In this study, we examined the distribution of various p120ctn isoforms and the consequences of their expression in cultured cells of epidermal origin. Immunohistochemical analysis and western blotting revealed that melanocytes and melanoma cells primarily express the long isoform 1A, whereas keratinocytes express shorter isoforms, especially 3A, which localize to cell-cell adhesion junctions in a calcium-dependent manner. The shortest isoform 4A, which was detected in normal keratinocytes and melanocytes, was generally lost from cells derived from squamous cell carcinomas or melanomas. The C-terminal alternatively spliced exon B was present in the p120ctn transcripts in the colon, intestine and prostate, but was lost in several tumor tissues derived from these organs. To test whether p120ctn isoforms serve in distinct biological functions, we transiently transfected the expression constructs into melanoma cells (1205-Lu) and immortalized keratinocytes (HaCaT). Indeed, distinct domains of p120ctn are responsible for its different biological functions. The prominent branching phenotype was induced equally by isoforms 1A, 2A and 3A, whereas the shortest isoform 4A, which was devoid of the N-terminal domain, completely lacked this ability. Also, the exon-B-encoded sequences, as in the isoform 1AB, were sufficient to abolish the branching phenotype as induced by the isoform 1A. The induction of the branching phenotype cosegregated with the nuclear localization of the p120ctn isoforms 1A, 2A and 3A, whereas the isoforms 4A and 1AB, which were excluded from the nucleus, did not induce the branching phenotype. The N-terminal sequences that contain seven out of eight tyrosine residues, recently characterized as potential candidates for phosphorylation by Src kinase, are required for the nuclear localization and for the formation of the branching phenotype. Finally, expression of the p120ctn isoforms, which caused the branching phenotype, was associated with cellular relocalization of E-cadherin in HaCaT cells. Collectively, we have identified sequences within the p120ctn N-terminus that are prerequisites for both nuclear localization and the p120ctn-induced branching phenotype. Loss of the cytoplasmic pool of p120ctn from tumor cells suggests an important function for such isoforms in normal cells and tissues.


Subject(s)
Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/chemistry , Cell Adhesion , Epithelium/chemistry , Neoplasms/chemistry , Phosphoproteins/analysis , Phosphoproteins/chemistry , Amino Acid Sequence , Animals , Cadherins/metabolism , Catenins , Cell Adhesion Molecules/genetics , Cell Communication , Cell Movement , Cells, Cultured , Cytoplasm/chemistry , Epithelium/ultrastructure , Humans , Infant, Newborn , Keratinocytes/metabolism , Melanocytes/metabolism , Mice , Models, Genetic , Mutation , Nuclear Localization Signals , Phosphoproteins/genetics , Protein Isoforms/analysis , Protein Isoforms/chemistry , Signal Transduction , Tumor Cells, Cultured , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...