Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Biol Evol ; 41(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38880992

ABSTRACT

Although evolution is driven by changes in how regulatory pathways control development, we know little about the molecular details underlying these transitions. The TRA-2 domain that mediates contact with TRA-1 is conserved in Caenorhabditis. By comparing the interaction of these proteins in two species, we identified a striking change in how sexual development is controlled. Identical mutations in this domain promote oogenesis in Caenorhabditis elegans but promote spermatogenesis in Caenorhabditis briggsae. Furthermore, the effects of these mutations involve the male-promoting gene fem-3 in C. elegans but are independent of fem-3 in C. briggsae. Finally, reciprocal mutations in these genes show that C. briggsae TRA-2 binds TRA-1 to prevent expression of spermatogenesis regulators. By contrast, in C. elegans TRA-1 sequesters TRA-2 in the germ line, allowing FEM-3 to initiate spermatogenesis. Thus, we propose that the flow of information within the sex determination pathway has switched directions during evolution. This result has important implications for how evolutionary change can occur.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Sex Determination Processes , Spermatogenesis , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Male , Spermatogenesis/genetics , Female , Caenorhabditis/genetics , Biological Evolution , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mutation , Oogenesis/genetics , Evolution, Molecular , Self-Fertilization , DNA-Binding Proteins , Transcription Factors
2.
Cell Commun Signal ; 20(1): 19, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177067

ABSTRACT

BACKGROUND: The Src tyrosine kinase phosphorylates effector proteins to induce expression of the podoplanin (PDPN) receptor in order to promote tumor progression. However, nontransformed cells can normalize the growth and morphology of neighboring transformed cells. Transformed cells must escape this process, called "contact normalization", to become invasive and malignant. Contact normalization requires junctional communication between transformed and nontransformed cells. However, specific junctions that mediate this process have not been defined. This study aimed to identify junctional proteins required for contact normalization. METHODS: Src transformed cells and oral squamous cell carcinoma cells were cultured with nontransformed cells. Formation of heterocellular adherens junctions between transformed and nontransformed cells was visualized by fluorescent microscopy. CRISPR technology was used to produce cadherin deficient and cadherin competent nontransformed cells to determine the requirement for adherens junctions during contact normalization. Contact normalization of transformed cells cultured with cadherin deficient or cadherin competent nontransformed cells was analyzed by growth assays, immunofluorescence, western blotting, and RNA-seq. In addition, Src transformed cells expressing PDPN under a constitutively active exogenous promoter were used to examine the ability of PDPN to override contact normalization. RESULTS: We found that N-cadherin (N-Cdh) appeared to mediate contact normalization. Cadherin competent cells that expressed N-Cdh inhibited the growth of neighboring transformed cells in culture, while cadherin deficient cells failed to inhibit the growth of these cells. Results from RNA-seq analysis indicate that about 10% of the transcripts affected by contact normalization relied on cadherin mediated communication, and this set of genes includes PDPN. In contrast, cadherin deficient cells failed to inhibit PDPN expression or normalize the growth of adjacent transformed cells. These data indicate that nontransformed cells formed heterocellular cadherin junctions to inhibit PDPN expression in adjacent transformed cells. Moreover, we found that PDPN enabled transformed cells to override the effects of contact normalization in the face of continued N-Cdh expression. Cadherin competent cells failed to normalize the growth of transformed cells expressing PDPN under a constitutively active exogenous promoter. CONCLUSIONS: Nontransformed cells form cadherin junctions with adjacent transformed cells to decrease PDPN expression in order to inhibit tumor cell proliferation. Cancer begins when a single cell acquires changes that enables them to form tumors. During these beginning stages of cancer development, normal cells surround and directly contact the cancer cell to prevent tumor formation and inhibit cancer progression. This process is called contact normalization. Cancer cells must break free from contact normalization to progress into a malignant cancer. Contact normalization is a widespread and powerful process; however, not much is known about the mechanisms involved in this process. This work identifies proteins required to form contacts between normal cells and cancer cells, and explores pathways by which cancer cells override contact normalization to progress into malignant cancers. Video Abstract.


Subject(s)
Antigens, CD , Cadherins , Mouth Neoplasms , Squamous Cell Carcinoma of Head and Neck , Adherens Junctions/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Transformation, Neoplastic , Humans , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
3.
Curr Biol ; 28(3): R121-R124, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29408260

ABSTRACT

A new study shows that the nematode Auanema rhodensis manipulates X chromosome segregation in surprising ways that depend on both the sex of the parent and the type of gamete. The result is a complex mating system that produces unusual sex ratios and inheritance patterns.


Subject(s)
Chromosome Segregation , Nematoda , Germ Cells , Humans , Male , Spermatozoa , X Chromosome
4.
Oncotarget ; 6(11): 9045-60, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25826087

ABSTRACT

Podoplanin (PDPN) is a unique transmembrane receptor that promotes tumor cell motility. Indeed, PDPN may serve as a chemotherapeutic target for primary and metastatic cancer cells, particularly oral squamous cell carcinoma (OSCC) cells that cause most oral cancers. Here, we studied how a monoclonal antibody (NZ-1) and lectin (MASL) that target PDPN affect human OSCC cell motility and viability. Both reagents inhibited the migration of PDPN expressing OSCC cells at nanomolar concentrations before inhibiting cell viability at micromolar concentrations. In addition, both reagents induced mitochondrial membrane permeability transition to kill OSCC cells that express PDPN by caspase independent nonapoptotic necrosis. Furthermore, MASL displayed a surprisingly robust ability to target PDPN on OSCC cells within minutes of exposure, and significantly inhibited human OSCC dissemination in zebrafish embryos. Moreover, we report that human OSCC cells formed tumors that expressed PDPN in mice, and induced PDPN expression in infiltrating host murine cancer associated fibroblasts. Taken together, these data suggest that antibodies and lectins may be utilized to combat OSCC and other cancers that express PDPN.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/pathology , Membrane Glycoproteins/antagonists & inhibitors , Molecular Targeted Therapy , Mouth Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Phytohemagglutinins/pharmacology , Administration, Oral , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Fibroblasts/pathology , Humans , Membrane Glycoproteins/immunology , Membrane Glycoproteins/physiology , Membrane Potential, Mitochondrial/drug effects , Mice , Mouth Neoplasms/virology , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Papillomaviridae/isolation & purification , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Phytohemagglutinins/administration & dosage , Phytohemagglutinins/therapeutic use , Xenograft Model Antitumor Assays , Zebrafish/embryology
5.
Mol Biol Evol ; 31(10): 2573-85, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24987105

ABSTRACT

The major families of chromatin remodelers have been conserved throughout eukaryotic evolution. Because they play broad, pleiotropic roles in gene regulation, it was not known if their functions could change rapidly. Here, we show that major alterations in the use of chromatin remodelers are possible, because the nucleosome remodeling factor (NURF) complex has acquired a unique role in the sperm/oocyte decision of the nematode Caenorhabditis briggsae. First, lowering the activity of C. briggsae NURF-1 or ISW-1, the core components of the NURF complex, causes germ cells to become oocytes rather than sperm. This observation is based on the analysis of weak alleles and null mutations that were induced with TALENs and on RNA interference. Second, qRT-polymerase chain reaction data show that the C. briggsae NURF complex promotes the expression of Cbr-fog-1 and Cbr-fog-3, two genes that control the sperm/oocyte decision. This regulation occurs in the third larval stage and affects the expression of later spermatogenesis genes. Third, double mutants reveal that the NURF complex and the transcription factor TRA-1 act independently on Cbr-fog-1 and Cbr-fog-3. TRA-1 binds both promoters, and computer analyses predict that these binding sites are buried in nucleosomes, so we suggest that the NURF complex alters chromatin structure to allow TRA-1 access to Cbr-fog-1 and Cbr-fog-3. Finally, lowering NURF activity by mutation or RNA interference does not affect this trait in other nematodes, including the sister species C. nigoni, so it must have evolved recently. We conclude that altered chromatin remodeling could play an important role in evolutionary change.


Subject(s)
Biological Evolution , Caenorhabditis/physiology , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Oocytes/metabolism , Spermatozoa/metabolism , Animals , Caenorhabditis/classification , Caenorhabditis/genetics , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Expression Regulation, Developmental , Male , Mutation , Sex Determination Processes , Species Specificity
6.
Mol Biol Evol ; 31(2): 468-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24194560

ABSTRACT

Although evolutionary studies of gene function often rely on RNA interference, the ideal approach would use reverse genetics to create null mutations for cross-species comparisons and forward genetics to identify novel genes in each species. We have used transcription activator-like effector nucleases (TALENs) to facilitate both approaches in Caenorhabditis nematodes. First, by combining golden gate cloning and TALEN technology, we can induce frameshifting mutations in any gene. Second, by combining this approach with bioinformatics we can predict and create the resources needed for forward genetic analysis in species like Caenorhabditis briggsae. Although developing genetic model organisms used to require years to isolate marker mutations, balancers, and tools, with TALENs, these reagents can now be produced in months. Furthermore, the analysis of nonsense mutants in related model organisms allows a directed approach for making these markers and tools. When used together, these methods could simplify the adaptation of other organisms for forward and reverse genetics.


Subject(s)
Caenorhabditis/genetics , Endonucleases/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , Biological Evolution , Caenorhabditis/metabolism , Cloning, Molecular , Computational Biology , Gene Knockout Techniques , Mutation , Species Specificity
7.
J Biol Chem ; 288(17): 12215-21, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23530051

ABSTRACT

Podoplanin (PDPN) is a transmembrane receptor that affects the activities of Rho, ezrin, and other proteins to promote tumor cell motility, invasion, and metastasis. PDPN is found in many types of cancer and may serve as a tumor biomarker and chemotherapeutic target. The intracellular region of PDPN contains only two serines, and these are conserved in mammals including mice and humans. We generated cells from the embryos of homozygous null Pdpn knock-out mice to investigate the relevance of these serines to cell growth and migration on a clear (PDPN-free) background. We report here that one or both of these serines can be phosphorylated by PKA (protein kinase A). We also report that conversion of these serines to nonphosphorylatable alanine residues enhances cell migration, whereas their conversion to phosphomimetic aspartate residues decreases cell migration. These results indicate that PKA can phosphorylate PDPN to decrease cell migration. In addition, we report that PDPN expression in fibroblasts causes them to facilitate the motility and viability of neighboring melanoma cells in coculture. These findings shed new light on how PDPN promotes cell motility, its role in tumorigenesis, and its utility as a functionally relevant biomarker and chemotherapeutic target.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line, Tumor , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/genetics , Fibroblasts/pathology , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phosphorylation/genetics , Serine/genetics , Serine/metabolism
8.
PLoS One ; 7(7): e41845, 2012.
Article in English | MEDLINE | ID: mdl-22844530

ABSTRACT

Cancer is a leading cause of death of men and women worldwide. Tumor cell motility contributes to metastatic invasion that causes the vast majority of cancer deaths. Extracellular receptors modified by α2,3-sialic acids that promote this motility can serve as ideal chemotherapeutic targets. For example, the extracellular domain of the mucin receptor podoplanin (PDPN) is highly O-glycosylated with α2,3-sialic acid linked to galactose. PDPN is activated by endogenous ligands to induce tumor cell motility and metastasis. Dietary lectins that target proteins containing α2,3-sialic acid inhibit tumor cell growth. However, anti-cancer lectins that have been examined thus far target receptors that have not been identified. We report here that a lectin from the seeds of Maackia amurensis (MASL) with affinity for O-linked carbohydrate chains containing sialic acid targets PDPN to inhibit transformed cell growth and motility at nanomolar concentrations. Interestingly, the biological activity of this lectin survives gastrointestinal proteolysis and enters the cardiovascular system to inhibit melanoma cell growth, migration, and tumorigenesis. These studies demonstrate how lectins may be used to help develop dietary agents that target specific receptors to combat malignant cell growth.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic , Membrane Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Plant Lectins/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Maackia/chemistry , Melanoma/blood supply , Melanoma/diet therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Molecular Sequence Data , Necrosis/chemically induced , Neovascularization, Pathologic/diet therapy , Plant Lectins/chemistry , Plant Lectins/metabolism , src-Family Kinases/metabolism
9.
J Biol Chem ; 285(13): 9649-9656, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20123990

ABSTRACT

Nontransformed cells can force tumor cells to assume a normal morphology and phenotype by the process of contact normalization. Transformed cells must escape this process to become invasive and malignant. However, mechanisms underlying contact normalization have not been elucidated. Here, we have identified genes that are affected by contact normalization of Src-transformed cells. Tumor cells must migrate to become invasive and malignant. Src must phosphorylate the adaptor protein Cas (Crk-associated substrate) to promote tumor cell motility. We report here that Src utilizes Cas to induce podoplanin (Pdpn) expression to promote tumor cell migration. Pdpn is a membrane-bound extracellular glycoprotein that associates with endogenous ligands to promote tumor cell migration leading to cancer invasion and metastasis. In fact, Pdpn expression accounted for a major part of the increased migration seen in Src-transformed cells. Moreover, nontransformed cells suppressed Pdpn expression in adjacent Src-transformed cells. Of >39,000 genes, Pdpn was one of only 23 genes found to be induced by transforming Src activity and suppressed by contact normalization of Src-transformed cells. In addition, we found 16 genes suppressed by Src and induced by contact normalization. These genes encode growth factor receptors, adaptor proteins, and products that have not yet been annotated and may play important roles in tumor cell growth and migration.


Subject(s)
Crk-Associated Substrate Protein/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/biosynthesis , src-Family Kinases/metabolism , Animals , Cell Line, Transformed , Cell Movement , Fibroblasts/metabolism , Homozygote , Ligands , Mice , Microscopy, Fluorescence/methods , Models, Biological , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/metabolism
10.
Oncotarget ; 1(3): 198-209, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21301049

ABSTRACT

Cell migration is an essential step in cancer invasion and metastasis. A number of orchestrated cellular events involving tyrosine kinases and signaling receptors enable cancer cells to dislodge from primary tumors and colonize elsewhere in the body. For example, activation of the Src and Abl kinases can mediate events that promote tumor cell migration. Also, activation of the Robo1 receptor can induce tumor cell migration. However, while the importance of Src, Abl, and Robo1 in cell migration have been demonstrated, molecular mechanisms by which they collectively influence cell migration have not been clearly elucidated. In addition, little is known about mechanisms that control Robo1 expression. We report here that Src activates Abl to stabilize Robo1 in order to promote cell migration. Inhibition of Abl kinase activity by siRNA or kinase blockers decreased Robo1 protein levels and suppressed the migration of transformed cells. We also provide evidence that Robo1 utilizes Cdc42 and Rac1 GTPases to induce cell migration. In addition, inhibition of Robo1 signaling can suppress transformed cell migration in the face of robust Src and Abl kinase activity. Therefore, inhibitors of Src, Abl, Robo1 and small GTPases may target a coordinated pathway required for tumor cell migration.


Subject(s)
Nerve Tissue Proteins/metabolism , Oncogene Proteins v-abl/metabolism , Receptors, Immunologic/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Microarray Analysis , Nerve Tissue Proteins/genetics , Oncogene Proteins v-abl/genetics , Protein Stability/drug effects , RNA, Small Interfering/genetics , Receptors, Immunologic/genetics , Transgenes/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Roundabout Proteins
11.
Cancer Sci ; 99(7): 1326-33, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18422756

ABSTRACT

The Src tyrosine kinase associates with the focal adhesion adaptor protein Cas (Crk-associated substrate) to suppress the expression of potential tumor suppressor genes. For example, Src utilizes Cas to suppress the expression of the LIM-only protein Fhl1 (four and a half LIM domains 1), in order to promote non-anchored tumor-cell growth and migration. Here, we report that the promoter region of the Fhl1 gene was methylated more in Src-transformed cells than non-transformed cells. In addition, global expression analysis indicates that Fhl1 induced expression of serum deprivation response factor (Sdpr) in Src-transformed cells. Moreover, Fhl1 and Sdpr was expressed in approximately 87% and 40% of samples obtained from non-transformed breast, 100% of samples obtained from non-transformed kidney, and over 60% of samples obtained from non-transformed prostate. In contrast, Fhl1 and Sdpr was detected in approximately 40% and 7% of matched samples from mammary carcinoma, less than 11% of matched samples from kidney carcinoma, and in less than 22% of matched samples from prostate carcinoma. These data indicate that Fhl1 and Sdpr expression was significantly reduced in tumors of the breast (P < 0.02 and P < 0.001), kidney (P < 0.01), and prostate (P < 0.05). In addition, although Src can activate mitogen-activated protein kinase (MAPK) to promote tumor-cell growth, our data indicate that Src did not rely on MAPK activity to suppress the expression of Fhl1 and Sdpr in transformed cells. Thus, Src induced methylation of the promoter region of the Fhl1 gene; Src suppressed Fhl1 and Sdpr expression independent of mitogen-activated protein kinase (MAPK) activity; Fhl1 induced the expression of Sdpr in Src-transformed cells; and Fhl1 and Sdpr expression was suppressed in tumors of the breast, kidney, and prostate.


Subject(s)
Breast Neoplasms/chemistry , Carrier Proteins/analysis , Intracellular Signaling Peptides and Proteins/analysis , Kidney Neoplasms/chemistry , Muscle Proteins/analysis , Prostatic Neoplasms/chemistry , Animals , Carrier Proteins/genetics , Cell Transformation, Neoplastic , Crk-Associated Substrate Protein/physiology , DNA Methylation , Disease Progression , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins , Male , Mice , Muscle Proteins/genetics , NIH 3T3 Cells , Phosphate-Binding Proteins , Promoter Regions, Genetic , src-Family Kinases/physiology
12.
J Biol Chem ; 282(26): 18914-21, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17488714

ABSTRACT

The Src tyrosine kinase phosphorylates Cas (Crk-associated substrate) to confer anchorage independence and invasive growth potential to transformed cells. Gap junctional communication is often lower between aggressive tumor cells compared with normal or benign precursors. The gap junction protein connexin43 (Cx43) is a tumor suppressor that can inhibit tumor cell growth. Src can phosphorylate Cx43 to block gap junctional communication between transformed cells. However, mechanisms by which this event actually closes intercellular channels have not been clearly defined. Here, we report that Src and Cas associate with each other at intercellular junctions. In addition, Cas is required for Src to reduce dye transfer and electrical coupling between cells expressing Cx43. Thus, Src utilizes Cas to inhibit gap junctional communication mediated by Cx43. This finding introduces a novel role of the Cas focal adhesion linker protein in the gap junction complex. This observation may help explain how gap junctional communication can be suppressed between malignant and metastatic tumor cells.


Subject(s)
Connexin 43/metabolism , Crk-Associated Substrate Protein/metabolism , Gap Junctions/metabolism , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism , Animals , Cell Communication/physiology , Cell Transformation, Neoplastic , Crk-Associated Substrate Protein/genetics , Focal Adhesions/metabolism , Mice , Mice, Knockout , Phosphorylation , RNA, Small Interfering , Tumor Suppressor Proteins/genetics , Tyrosine/metabolism
13.
J Biol Chem ; 281(30): 20689-20697, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16707485

ABSTRACT

Cas is a multidomain signaling protein that resides in focal adhesions. Cas possesses a large central substrate domain containing 15 repeats of the sequence YXXP, which are phosphorylated by Src. The phosphorylation sites are essential for the roles of Cas in cell migration and in regulation of the actin cytoskeleton. We showed previously that Src catalyzes the multisite phosphorylation of Cas via a processive mechanism. In this study, we created mutant forms of Cas to identify the determinants for processive phosphorylation. Mutants containing single or multiple YXXP mutations were phosphorylated processively by Src, suggesting that individual sites are dispensable. The results also suggest that there is no defined order to the Cas phosphorylation events. We also studied the effects of these mutations by reintroducing Cas into Cas-deficient fibroblasts. Mutants lacking some or all YXXP sites augment the ability of Src to promote anchorage-independent growth. On the other hand, deletion of YXXP sites compromises the ability of Cas to promote tumor cell migration.


Subject(s)
Crk-Associated Substrate Protein/chemistry , src-Family Kinases/chemistry , Animals , Binding Sites , Cell Movement , Fibroblasts/metabolism , Insecta , Kinetics , Mice , Models, Genetic , Mutagenesis, Site-Directed , Mutation , Neoplasms/metabolism , Phosphorylation , src-Family Kinases/metabolism
14.
Cancer Res ; 66(3): 1543-52, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452211

ABSTRACT

Anchorage independence and motility are hallmarks of tumor cell growth. Tumor cell growth and morphology can be normalized by contact with nontransformed cells. The Src tyrosine kinase phosphorylates specific sites on the focal adhesion adaptor protein Crk-associated substrate (Cas) to promote nonanchored cell growth and migration. We studied the effects of Src and Cas on the expression of >14,000 genes to identify molecular events that underlie these activities. Gene expression in tumor cells that were normalized by neighboring nontransformed cells was used as an additional filter to identify genes that control metastatic cell growth. This process enabled the identification of genes that play roles in anchorage-independent cell growth and migration. One candidate, four and a half LIM domains 1 (Fhl1), acts as a transcriptional regulator that can associate with cell junctions as well as with the nucleus. We show here that Src phosphorylates Cas to block Fhl1 expression. In addition, suppression of Fhl1 is required for Src to promote tumor cell growth. These data show that Fhl1 is a tumor suppressor gene that acts downstream of Src and Cas to specifically block anchorage-independent cell growth and migration. Moreover, Fhl1 was suppressed in tumors from several human tissues. Thus, identification of how Fhl1 controls fundamental aspects of tumor cell growth and metastasis may lead to the development of novel markers that can be used to diagnose human clinical specimens as well as open innovative avenues of investigations aimed at developing reagents that target cancer cells while minimizing damage to normal cells.


Subject(s)
Cell Movement/physiology , Cellular Apoptosis Susceptibility Protein/metabolism , Muscle Proteins/antagonists & inhibitors , Neoplasms, Experimental/pathology , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Growth Processes/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...