Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
2.
Elife ; 132024 May 07.
Article in English | MEDLINE | ID: mdl-38712822

ABSTRACT

Pancreatic ductal adenocarcinoma carries a dismal prognosis, with high rates of metastasis and few treatment options. Hyperactivation of KRAS in almost all tumours drives RAC1 activation, conferring enhanced migratory and proliferative capacity as well as macropinocytosis. Macropinocytosis is well understood as a nutrient scavenging mechanism, but little is known about its functions in trafficking of signalling receptors. We find that CYRI-B is highly expressed in pancreatic tumours in a mouse model of KRAS and p53-driven pancreatic cancer. Deletion of Cyrib (the gene encoding CYRI-B protein) accelerates tumourigenesis, leading to enhanced ERK and JNK-induced proliferation in precancerous lesions, indicating a potential role as a buffer of RAC1 hyperactivation in early stages. However, as disease progresses, loss of CYRI-B inhibits metastasis. CYRI-B depleted tumour cells show reduced chemotactic responses to lysophosphatidic acid, a major driver of tumour spread, due to impaired macropinocytic uptake of the lysophosphatidic acid receptor 1. Overall, we implicate CYRI-B as a mediator of growth and signalling in pancreatic cancer, providing new insights into pathways controlling metastasis.


Pancreatic cancer is an aggressive disease with limited treatment options. It is also associated with high rates of metastasis ­ meaning it spreads to other areas of the body. Environmental pressures, such as a lack of the nutrients metastatic cancer cells need to grow and divide, can change how the cells behave. Understanding the changes that allow cancer cells to respond to these pressures could reveal new treatment options for pancreatic cancer. When nutrients are scarce, metastatic cancer cells can gather molecules and nutrients by capturing large amounts of the fluid that surrounds them using a mechanism called macropinocytosis. They can also migrate to areas of the body with higher nutrient levels, through a process called chemotaxis. This involves cells moving towards areas with higher levels of certain molecules. For example, cancer cells migrate towards high levels of a lipid called lysophosphatidic acid, which promotes their growth and survival. A newly discovered protein known as CYRI-B has recently been shown to regulate how cells migrate and take up nutrients. It also interacts with proteins known to be involved in pancreatic cancer progression. Therefore, Nikolaou et al. set out to investigate whether CYRI-B also plays a role in metastatic pancreatic cancer. Experiments in a mouse model of pancreatic cancer showed that CYRI-B levels were high in pancreatic tumour cells. And when the gene for CYRI-B was removed from the tumour cells, they did not metastasise. Further analysis revealed that CYRI-B controls uptake and processing of nutrients and other signalling molecules through macropinocytosis. In particular, it ensures uptake of the receptor for lysophosphatidic acid, allowing the metastatic cancer cells to migrate. The findings of Nikolaou et al. reveal that CYRI-B is involved in metastasis of cancer cells in a mouse model of pancreatic cancer. This new insight into how metastasis is controlled could help to identify future targets for treatments that aim to prevent pancreatic cancer cells spreading to distant sites.


Subject(s)
Pancreatic Neoplasms , Pinocytosis , Receptors, Lysophosphatidic Acid , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics
3.
Sci Signal ; 15(720): eabd9099, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35133863

ABSTRACT

Genetically encoded probes are widely used to visualize cellular processes in vitro and in vivo. Although effective in cultured cells, fluorescent protein tags and reporters are suboptimal in vivo because of poor tissue penetration and high background signal. Luciferase reporters offer improved signal-to-noise ratios but require injections of luciferin that can lead to variable responses and that limit the number and timing of data points that can be gathered. Such issues in studying the critical transcription factor p53 have limited insight on its activity in vivo during development and tissue injury responses. Here, by linking the expression of the near-infrared fluorescent protein iRFP713 to a synthetic p53-responsive promoter, we generated a knock-in reporter mouse that enabled noninvasive, longitudinal analysis of p53 activity in vivo in response to various stimuli. In the developing embryo, this model revealed the timing and localization of p53 activation. In adult mice, the model monitored p53 activation in response to irradiation and paracetamol- or CCl4-induced liver regeneration. After irradiation, we observed potent and sustained activation of p53 in the liver, which limited the production of reactive oxygen species (ROS) and promoted DNA damage resolution. We propose that this new reporter may be used to further advance our understanding of various physiological and pathophysiological p53 responses.


Subject(s)
Liver Regeneration , Tumor Suppressor Protein p53 , Animals , DNA Damage , Genes, Reporter , Liver Regeneration/genetics , Mice , Promoter Regions, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Elife ; 102021 04 19.
Article in English | MEDLINE | ID: mdl-33871359

ABSTRACT

Apoptosis is characterized by profound morphological changes, but their physiological purpose is unknown. To characterize the role of apoptotic cell contraction, ROCK1 was rendered caspase non-cleavable (ROCK1nc) by mutating aspartate 1113, which revealed that ROCK1 cleavage was necessary for forceful contraction and membrane blebbing. When homozygous ROCK1nc mice were treated with the liver-selective apoptotic stimulus of diethylnitrosamine, ROCK1nc mice had more profound liver damage with greater neutrophil infiltration than wild-type mice. Inhibition of the damage-associated molecular pattern protein HMGB1 or signalling by its cognate receptor TLR4 lowered neutrophil infiltration and reduced liver damage. ROCK1nc mice also developed fewer diethylnitrosamine-induced hepatocellular carcinoma (HCC) tumours, while HMGB1 inhibition increased HCC tumour numbers. Thus, ROCK1 activation and consequent cell contraction are required to limit sterile inflammation and damage amplification following tissue-scale cell death. Additionally, these findings reveal a previously unappreciated role for acute sterile inflammation as an efficient tumour-suppressive mechanism.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/prevention & control , Cell Shape , Chemical and Drug Induced Liver Injury/pathology , Liver Neoplasms/prevention & control , Liver/pathology , rho-Associated Kinases/metabolism , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Caspases/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/etiology , Diethylnitrosamine , Disease Models, Animal , Enzyme Activation , Glycyrrhizic Acid , HEK293 Cells , HMGB1 Protein/metabolism , Humans , Liver/enzymology , Liver Neoplasms/chemically induced , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Mutation , Myosin Light Chains/metabolism , Neutrophil Infiltration , Phosphorylation , Sulfonamides , Toll-Like Receptor 4/metabolism , rho-Associated Kinases/genetics
6.
Oncogene ; 39(8): 1797-1806, 2020 02.
Article in English | MEDLINE | ID: mdl-31740786

ABSTRACT

BRF1 is a rate-limiting factor for RNA Polymerase III-mediated transcription and is elevated in numerous cancers. Here, we report that elevated levels of BRF1 associate with poor prognosis in human prostate cancer. In vitro studies in human prostate cancer cell lines demonstrated that transient overexpression of BRF1 increased cell proliferation whereas the transient downregulation of BRF1 reduced proliferation and mediated cell cycle arrest. Consistent with our clinical observations, BRF1 overexpression in a Pten-deficient mouse (PtenΔ/Δ BRF1Tg) prostate cancer model accelerated prostate carcinogenesis and shortened survival. In PtenΔ/Δ BRF1Tg tumours, immune and inflammatory processes were altered, with reduced tumoral infiltration of neutrophils and CD4 positive T cells, which can be explained by decreased levels of complement factor D (CFD) and C7 components of the complement cascade, an innate immune pathway that influences the adaptive immune response. We tested if the secretome was involved in BRF1-driven tumorigenesis. Unbiased proteomic analysis on BRF1-overexpresing PC3 cells confirmed reduced levels of CFD in the secretome, implicating the complement system in prostate carcinogenesis. We further identify that expression of C7 significantly correlates with expression of CD4 and has the potential to alter clinical outcome in human prostate cancer, where low levels of C7 associate with poorer prognosis.


Subject(s)
Carcinogenesis , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , TATA-Binding Protein Associated Factors/metabolism , Aged , CD4-Positive T-Lymphocytes/immunology , Cell Cycle , Cell Proliferation , Humans , Male , Middle Aged , PTEN Phosphohydrolase/metabolism , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism
7.
Nat Cell Biol ; 21(11): 1413-1424, 2019 11.
Article in English | MEDLINE | ID: mdl-31685988

ABSTRACT

Tumours depend on altered rates of protein synthesis for growth and survival, which suggests that mechanisms controlling mRNA translation may be exploitable for therapy. Here, we show that loss of APC, which occurs almost universally in colorectal tumours, strongly enhances the dependence on the translation initiation factor eIF2B5. Depletion of eIF2B5 induces an integrated stress response and enhances translation of MYC via an internal ribosomal entry site. This perturbs cellular amino acid and nucleotide pools, strains energy resources and causes MYC-dependent apoptosis. eIF2B5 limits MYC expression and prevents apoptosis in APC-deficient murine and patient-derived organoids and in APC-deficient murine intestinal epithelia in vivo. Conversely, the high MYC levels present in APC-deficient cells induce phosphorylation of eIF2α via the kinases GCN2 and PKR. Pharmacological inhibition of GCN2 phenocopies eIF2B5 depletion and has therapeutic efficacy in tumour organoids, which demonstrates that a negative MYC-eIF2α feedback loop constitutes a targetable vulnerability of colorectal tumours.


Subject(s)
Colorectal Neoplasms/genetics , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/antagonists & inhibitors , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Feedback, Physiological , Female , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays
8.
Blood ; 134(25): 2318-2329, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31697813

ABSTRACT

During platelet spreading, the actin cytoskeleton undergoes rapid rearrangement, forming filopodia and lamellipodia. Controversial data have been published on the role of lamellipodia in thrombus formation and stability. The Wiskott-Aldrich syndrome protein-family verprolin-homologous protein (WAVE)-regulatory complex, which has been shown in other cells to drive lamellipodium formation by enhancing actin nucleation via the actin-related protein 2/3 (Arp2/3) complex, is activated by Ras-related C3 botulinum toxin substrate 1 (Rac1) interaction with the WAVE complex subunit cytoplasmic fragile X mental retardation 1-interacting protein 1 (Cyfip1). We analyzed Cyfip1flox/floxPf4-Cre mice to investigate the role of Cyfip1 in platelet function. These mice displayed normal platelet counts and a slight reduction in platelet volume. Activation of mutant platelets was only moderately reduced to all tested agonists as measured by αIIbß3 integrin activation and P-selectin surface exposure. However, lamellipodium formation of mutant platelets was completely abolished on different matrices. Nevertheless, Cyfip1-deficient platelets formed stable thrombi on collagen fibers ex vivo and in 2 models of occlusive arterial thrombosis in vivo. Similarly, the hemostatic function and maintenance of vascular integrity during inflammation of the skin and lung were unaltered in the mutant mice. Investigation of platelet morphology in an induced thrombus under flow revealed that platelets rather form filopodia in the thrombus shell, and are flattened with filopodium-like structures when in direct contact to collagen fibers at the bottom of the thrombus. We provide for the first time direct evidence that platelet lamellipodium formation is not required for stable thrombus formation, and that morphological changes of platelets differ between a static spreading assay and thrombus formation under flow.


Subject(s)
Blood Platelets/metabolism , Pseudopodia/metabolism , Thrombosis/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Platelets/pathology , Female , Male , Mice , Mice, Transgenic , Neuropeptides/genetics , Neuropeptides/metabolism , P-Selectin/genetics , P-Selectin/metabolism , Pseudopodia/genetics , Thrombosis/genetics , Thrombosis/pathology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
9.
Cell Death Differ ; 26(12): 2535-2550, 2019 12.
Article in English | MEDLINE | ID: mdl-30858608

ABSTRACT

RNA polymerase III (Pol-III) transcribes tRNAs and other small RNAs essential for protein synthesis and cell growth. Pol-III is deregulated during carcinogenesis; however, its role in vivo has not been studied. To address this issue, we manipulated levels of Brf1, a Pol-III transcription factor that is essential for recruitment of Pol-III holoenzyme at tRNA genes in vivo. Knockout of Brf1 led to embryonic lethality at blastocyst stage. In contrast, heterozygous Brf1 mice were viable, fertile and of a normal size. Conditional deletion of Brf1 in gastrointestinal epithelial tissues, intestine, liver and pancreas, was incompatible with organ homeostasis. Deletion of Brf1 in adult intestine and liver induced apoptosis. However, Brf1 heterozygosity neither had gross effects in these epithelia nor did it modify tumorigenesis in the intestine or pancreas. Overexpression of BRF1 rescued the phenotypes of Brf1 deletion in intestine and liver but was unable to initiate tumorigenesis. Thus, Brf1 and Pol-III activity are absolutely essential for normal homeostasis during development and in adult epithelia. However, Brf1 overexpression or heterozygosity are unable to modify tumorigenesis, suggesting a permissive, but not driving role for Brf1 in the development of epithelial cancers of the pancreas and gut.


Subject(s)
Butyrate Response Factor 1/deficiency , Intestinal Mucosa/metabolism , Liver/metabolism , Pancreas/metabolism , Animals , Butyrate Response Factor 1/biosynthesis , Butyrate Response Factor 1/genetics , Homeostasis , Humans , Mice , TATA-Binding Protein Associated Factors/biosynthesis , TATA-Binding Protein Associated Factors/genetics
10.
Genesis ; 54(12): 636-646, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27775859

ABSTRACT

The serine/threonine kinases ROCK1 and ROCK2 are central mediators of actomyosin contractile force generation that act downstream of the RhoA small GTP-binding protein. As a result, they have key roles in regulating cell morphology and proliferation, and have been implicated in numerous pathological conditions and diseases including hypertension and cancer. Here we describe the generation of a gene-targeted mouse line that enables CRE-inducible expression of a conditionally-active fusion between the ROCK2 kinase domain and the hormone-binding domain of a mutated estrogen receptor (ROCK2:ER). This two-stage system of regulation allows for tissue-selective expression of the ROCK2:ER fusion protein, which then requires administration of estrogen analogues such as tamoxifen or 4-hydroxytamoxifen to elicit kinase activity. This conditional gain-of-function system was validated in multiple tissues by crossing with mice expressing CRE recombinase under the transcriptional control of cytokeratin14 (K14), murine mammary tumor virus (MMTV) or cytochrome P450 Cyp1A1 (Ah) promoters, driving appropriate expression in the epidermis, mammary or intestinal epithelia respectively. Given the interest in ROCK signaling in normal physiology and disease, this mouse line will facilitate research into the consequences of ROCK activation that could be used to complement conditional knockout models. Birth Defects Research (Part A) 106:636-646, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Estrogen Receptor alpha/genetics , Recombinant Fusion Proteins/genetics , rho-Associated Kinases/genetics , Animals , Cytochrome P-450 CYP1A1/genetics , Epidermis/metabolism , Estrogen Receptor alpha/biosynthesis , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Integrases/genetics , Intestinal Mucosa/metabolism , Mammary Glands, Animal/metabolism , Mice , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/drug effects , Tamoxifen/administration & dosage , rho-Associated Kinases/biosynthesis
11.
Cancer Cell ; 29(6): 832-845, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27265504

ABSTRACT

CXCR2 has been suggested to have both tumor-promoting and tumor-suppressive properties. Here we show that CXCR2 signaling is upregulated in human pancreatic cancer, predominantly in neutrophil/myeloid-derived suppressor cells, but rarely in tumor cells. Genetic ablation or inhibition of CXCR2 abrogated metastasis, but only inhibition slowed tumorigenesis. Depletion of neutrophils/myeloid-derived suppressor cells also suppressed metastasis suggesting a key role for CXCR2 in establishing and maintaining the metastatic niche. Importantly, loss or inhibition of CXCR2 improved T cell entry, and combined inhibition of CXCR2 and PD1 in mice with established disease significantly extended survival. We show that CXCR2 signaling in the myeloid compartment can promote pancreatic tumorigenesis and is required for pancreatic cancer metastasis, making it an excellent therapeutic target.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Receptors, Interleukin-8B/genetics , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Mice , Neoplasm Metastasis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prognosis , Receptors, Interleukin-8B/antagonists & inhibitors , Signal Transduction , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , Survival Analysis , Up-Regulation , Xenograft Model Antitumor Assays , Gemcitabine
12.
PLoS One ; 10(6): e0131066, 2015.
Article in English | MEDLINE | ID: mdl-26114544

ABSTRACT

Barth syndrome is an X-linked mitochondrial disease, symptoms of which include neutropenia and cardiac myopathy. These symptoms are the most significant clinical consequences of a disease, which is increasingly recognised to have a variable presentation. Mutation in the Taz gene in Xq28 is thought to be responsible for the condition, by altering mitochondrial lipid content and mitochondrial function. Male chimeras carrying a targeted mutation of Taz on their X-chromosome were infertile. Testes from the Taz knockout chimeras were smaller than their control counterparts and this was associated with a disruption of the progression of spermatocytes through meiosis to spermiogenesis. Taz knockout ES cells also showed a defect when differentiated to germ cells in vitro. Mutant spermatocytes failed to progress past the pachytene stage of meiosis and had higher levels of DNA double strand damage and increased levels of endogenous retrotransposon activity. Altogether these data revealed a novel role for Taz in helping to maintain genome integrity in meiosis and facilitating germ cell differentiation. We have unravelled a novel function for the Taz protein, which should contribute to an understanding of how a disruption of the Taz gene results in the complex symptoms underlying Barth Syndrome.


Subject(s)
Meiosis/genetics , Spermatocytes/physiology , Spermatogenesis/genetics , Transcription Factors/physiology , Acyltransferases , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/physiology , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics
13.
Genesis ; 47(7): 440-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19391117

ABSTRACT

ROCK kinases regulate actin-myosin structures downstream of Rho GTPases. We generated mice expressing 4-hydroxytamoxifen (4HT)-regulated human ROCK II (ROCKII:mER) under the transcriptional control of the cytokeratin14 (K14) promoter. The K14-ROCKII:mER minigene was recombineered into a novel cloning vector containing the promoter and first exon of the human HPRT gene, and second and third exons of the mouse Hprt gene. Homologous recombination into the Hprt locus, which is deleted for the promoter and first two exons in HM1 embryonic stem cells, reconstitutes a functional Hprt gene, allowing for growth in HAT (hypoxanthine-aminopterin-thymidine) medium. K14-promoter-driven ROCKII:mER expression was restricted to a superficial cell layer in embryoid bodies, with increased ROCK substrate phosphorylation induced by 4HT. ROCKII:mER-expressing primary murine keratinocytes responded to 4HT with increased substrate phosphorylation and cytoskeleton rearrangements, indicating that ROCKII:mER activity is regulated by 4HT in the target tissue. K14-ROCKII:mER mice will be valuable for examining the role of ROCK in skin development and cancer.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Targeting , rho-Associated Kinases/genetics , Animals , Base Sequence , Cells, Cultured , DNA Primers , Embryonic Stem Cells/enzymology , Exons , Genetic Vectors , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Immunohistochemistry , Keratin-14/genetics , Mice , Polymerase Chain Reaction , Promoter Regions, Genetic , Recombination, Genetic , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
14.
Nature ; 432(7018): 775-9, 2004 Dec 09.
Article in English | MEDLINE | ID: mdl-15592418

ABSTRACT

The FBXW7/hCDC4 gene encodes a ubiquitin ligase implicated in the control of chromosome stability. Here we identify the mouse Fbxw7 gene as a p53-dependent tumour suppressor gene by using a mammalian genetic screen for p53-dependent genes involved in tumorigenesis. Radiation-induced lymphomas from p53+/- mice, but not those from p53-/- mice, show frequent loss of heterozygosity and a 10% mutation rate of the Fbxw7 gene. Fbxw7+/- mice have greater susceptibility to radiation-induced tumorigenesis, but most tumours retain and express the wild-type allele, indicating that Fbxw7 is a haploinsufficient tumour suppressor gene. Loss of Fbxw7 alters the spectrum of tumours that develop in p53 deficient mice to include a range of tumours in epithelial tissues such as the lung, liver and ovary. Mouse embryo fibroblasts from Fbxw7-deficient mice, or wild-type mouse cells expressing Fbxw7 small interfering RNA, have higher levels of Aurora-A kinase, c-Jun and Notch4, but not of cyclin E. We propose that p53-dependent loss of Fbxw7 leads to genetic instability by mechanisms that might involve the activation of Aurora-A, providing a rationale for the early occurrence of these mutations in human cancers.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Genes, Tumor Suppressor/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Neoplastic/radiation effects , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Fibroblasts , Gene Deletion , Loss of Heterozygosity/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Survival Rate , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
15.
Cell Commun Adhes ; 10(4-6): 359-64, 2003.
Article in English | MEDLINE | ID: mdl-14681042

ABSTRACT

To elucidate the mode of action of dominant mutant connexins in causing inherited skin diseases, transgenic mice were produced that express the true Vohwinkel syndrome-associated mutant Cx26 (D66H), from a keratin 10 promoter, specifically in the suprabasal epidermal keratinocytes. Following birth, the transgenic mice developed keratoderma similar to that of human carriers of Cx26 (D66H). Expression of the transgene resulted in a loss of Cx26 and Cx30 at intercellular junctions of epidermal keratinocytes and accumulation of these connexins in the cytoplasm. Injection of primary mouse keratinocytes with Lucifer Yellow showed no difference in terms of dye spreading between transgenic and non transgenic keratinocytes in vitro. Expression of the mutant Cx26 (D66H) did not interfere with the formation of the epidermal water barrier during late embryonic development. Attempts to produce transgenic mice expressing the wild type form of Cx26 from the K10 promoter failed to produce viable animals although transgenic embryos were recovered at days 9 and 12 of gestation, suggesting that the transgene might be embryonic lethal.


Subject(s)
Cell Communication/physiology , Connexins/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Animals , Cell Communication/genetics , Connexin 26 , Connexin 30 , Connexins/genetics , Embryo, Mammalian/metabolism , Epidermis/embryology , Genes, Dominant , Isoquinolines/chemistry , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics
16.
Hum Mol Genet ; 12(14): 1737-44, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12837696

ABSTRACT

To investigate the role of connexins in dominantly inherited skin disease, transgenic mice were produced which expressed mutant connexin 26 [gjb2/connexin 26(D66H)], from a keratin 10 promoter, exclusively in the suprabasal epidermis (the cells in which Connexin 26 is up-regulated in epidermal hyperproliferative states). From soon after birth, the mice exhibited a keratoderma similar to that in humans carrying the Connexin 26(D66H) mutation (true Vohwinkel syndrome). Transgene expression was associated with loss of Connexin 26 and Connexin 30 from epidermal keratinocyte intercellular junctions and accumulation in cytoplasm. Light and electron microscopy showed marked thickening of the epidermal cornified layers and increased epidermal TUNEL staining, indicative of premature keratinocyte programmed cell death. The K10Connexin 26(D66H) mouse may provide a valuable model to study the role of gap-junctional intercellular communication in epidermal differentiation. Similarities in phenotype between individuals (man and mouse) carrying Connexin 26(D66H) and those carrying insertional mutants of Loricrin, a major cornified envelope protein of the epidermis, suggest a possible link between connexin function and cornified envelope formation.


Subject(s)
Connexins/genetics , Epidermis/metabolism , Genes, Dominant , Skin Abnormalities/genetics , Animals , Connexin 26 , Connexins/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL