Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunol Cell Biol ; 98(8): 626-638, 2020 09.
Article in English | MEDLINE | ID: mdl-32479655

ABSTRACT

Psoriasis (PS) and atopic dermatitis (AD) are common inflammatory skin diseases characterized by an imbalance in specific T-cell subsets, resulting in a specific cytokine profile in patients. Obtaining models closely resembling both pathologies along with a relevant clinical impact is crucial for the development of new therapies because of the high prevalence of these diseases. Single-gene mouse models developed until now do not fully reflect the complexity of these disorders, in part not only because of inherent differences between mice and humans but also because of the multifactorial nature of these pathologies. The skin-humanized mouse model developed by our group, based on a tissue engineering approach, has been used to test therapeutic strategies, although this methodology is still technically challenging and not widely available. The skin-humanized mouse models for PS and AD reproduce human skin phenotypes, providing valuable tools for drug development and testing in the preclinical setting. The tissue engineering approach allows the development of personalized medicine, covering the broad genotypic spectrum of these pathologies. This review highlights the main differences between available murine models focusing on the tissue-specific immunity of PS and AD. We discuss their contribution to unravel the complex pathophysiology of these diseases and to translate this knowledge into more accurate therapies.


Subject(s)
Dermatitis, Atopic , Disease Models, Animal , Immunity , Psoriasis , Animals , Cytokines , Dermatitis, Atopic/immunology , Humans , Mice , Psoriasis/immunology , Skin , T-Lymphocyte Subsets
2.
Int J Mol Sci ; 21(6)2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32178458

ABSTRACT

The role of stroma is fundamental in the development and behavior of epithelial tumors. In this regard, limited growth of squamous cell carcinomas (SCC) or cell-lines derived from them has been achieved in immunodeficient mice. Moreover, lack of faithful recapitulation of the original human neoplasia complexity is often observed in xenografted tumors. Here, we used tissue engineering techniques to recreate a humanized tumor stroma for SCCs grafted in host mice, by combining CAF (cancer associated fibroblasts)-like cells with a biocompatible scaffold. The stroma was either co-injected with epithelial cell lines derived from aggressive SCC or implanted 15 days before the injection of the tumoral cells, to allow its vascularization and maturation. None of the mice injected with the cell lines without stroma were able to develop a SCC. In contrast, tumors were able to grow when SCC cells were injected into previously established humanized stroma. Histologically, all of the regenerated tumors were moderately differentiated SCC with a well-developed stroma, resembling that found in the original human neoplasm. Persistence of human stromal cells was also confirmed by immunohistochemistry. In summary, we provide a proof of concept that humanized tumor stroma, generated by tissue engineering, can facilitate the development of epithelial tumors in immunodeficient mice.


Subject(s)
Carcinoma, Squamous Cell/pathology , Heterografts/pathology , Neoplasm Transplantation/pathology , Stromal Cells/pathology , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line , Cell Line, Tumor , Epithelial Cells/pathology , Female , Fibroblasts/pathology , Humans , Mice , Neovascularization, Pathologic/pathology , Tissue Engineering/methods , Transplantation, Heterologous/methods
3.
Exp Dermatol ; 25(4): 269-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26739954

ABSTRACT

Epidermolysis bullosa with pyloric atresia (EB-PA) is a rare autosomal recessive hereditary disease with a variable prognosis from lethal to very mild. EB-PA is classified into Simplex form (EBS-PA: OMIM #612138) and Junctional form (JEB-PA: OMIM #226730), and it is caused by mutations in ITGA6, ITGB4 and PLEC genes. We report the analysis of six patients with EB-PA, including two dizygotic twins. Skin immunofluorescence epitope mapping was performed followed by PCR and direct sequencing of the ITGB4 gene. Two of the patients presented with non-lethal EB-PA associated with missense ITGB4 gene mutations. For the other four, early postnatal demise was associated with complete lack of ß4 integrin due to a variety of ITGB4 novel mutations (2 large deletions, 1 splice-site mutation and 3 missense mutations). One of the deletions spanned 278 bp, being one of the largest reported to date for this gene. Remarkably, we also found for the first time a founder effect for one novel mutation in the ITGB4 gene. We have identified 6 novel mutations in the ITGB4 gene to be added to the mutation database. Our results reveal genotype-phenotype correlations that contribute to the molecular understanding of this heterogeneous disease, a pivotal issue for prognosis and for the development of novel evidence-based therapeutic options for EB management.


Subject(s)
Ectodermal Dysplasia/genetics , Integrin beta4/genetics , Sequence Deletion , Biopsy , Child, Preschool , DNA Mutational Analysis , Ectodermal Dysplasia/diagnosis , Epitope Mapping , Epitopes/chemistry , Female , Genetic Association Studies , Humans , Infant , Infant, Newborn , Keratinocytes/cytology , Male , Microsatellite Repeats/genetics , Microscopy, Fluorescence , Mutation, Missense , Polymerase Chain Reaction , Prognosis , Sequence Analysis, DNA , Twins, Dizygotic
4.
Mol Carcinog ; 52(6): 446-58, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22290600

ABSTRACT

The prevalence of obesity, an established risk and progression factor for postmenopausal breast cancer, remains high in US women. Activation of Akt/mammalian target of rapamycin (mTOR) signaling plays a key role in the obesity-breast cancer link. However, the impact of weight normalization in obese postmenopausal women on breast tumorigenesis and/or Akt/mTOR activation is poorly characterized. To model this, ovariectomized female C57BL/6 mice were fed a control diet (n = 20), a calorie restriction (CR) regimen (n = 20), or a diet-induced obesity (DIO) diet (n = 30). At week 17, DIO mice were switched to control diet, resulting in formerly obese (FOb) mice with weights identical to the controls by week 20. MMTV-Wnt-1 mammary tumor cells were injected at 20 wk into each mouse. Two weeks post-injection, vehicle or the mTOR inhibitor RAD001 at 10 or 15 mg/kg body weight (n = 10/diet group) was administered by gavage twice/week until termination. Relative to controls, CR mice had decreased (and DIO mice had increased) serum insulin-like growth factor-1 (IGF-1) and phosphorylation of Akt/mTOR pathway components. RAD001 decreased tumor growth in the CR, control, and FOb mice. Wnt-1 tumor cells treated in vitro with serum from mice from each group established that diet-dependent circulating factors contribute to tumor growth and invasiveness. These findings suggest weight normalization in obese mice does not immediately reverse tumor progression or Akt/mTOR activation. Treatment with RAD001 blocked mammary tumor development and mTOR activation observed in the FOb mice, suggesting combination of lifestyle and pharmacologic strategies may be effective for breaking the obesity-breast cancer link.


Subject(s)
Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/drug therapy , Obesity/complications , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Diet , Everolimus , Female , Hormones/blood , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Signal Transduction/drug effects , Sirolimus/therapeutic use , Weight Loss/drug effects , Wnt1 Protein/metabolism
5.
Exp Dermatol ; 22(3): 195-201, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23489422

ABSTRACT

Cutaneous diabetic wounds greatly affect the quality of life of patients, causing a substantial economic impact on the healthcare system. The limited clinical success of conventional treatments is mainly attributed to the lack of knowledge of the pathogenic mechanisms related to chronic ulceration. Therefore, management of diabetic ulcers remains a challenging clinical issue. Within this context, reliable animal models that recapitulate situations of impaired wound healing have become essential. In this study, we established a new in vivo humanised model of delayed wound healing in a diabetic context that reproduces the main features of the human disease. Diabetes was induced by multiple low doses of streptozotocin in bioengineered human-skin-engrafted immunodeficient mice. The significant delay in wound closure exhibited in diabetic wounds was mainly attributed to alterations in the granulation tissue formation and resolution, involving defects in wound bed maturation, vascularisation, inflammatory response and collagen deposition. In the new model, a cell-based wound therapy consisting of the application of plasma-derived fibrin dermal scaffolds containing fibroblasts consistently improved the healing response by triggering granulation tissue maturation and further providing a suitable matrix for migrating keratinocytes during wound re-epithelialisation. The present preclinical wound healing model was able to shed light on the biological processes responsible for the improvement achieved, and these findings can be extended for designing new therapeutic approaches with clinical relevance.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Fibroblasts/physiology , Regeneration/physiology , Skin Physiological Phenomena , Wound Healing/physiology , Animals , Bioengineering/methods , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Female , Fibroblasts/cytology , Humans , Mice , Mice, Nude , Streptozocin/adverse effects , Time Factors , Tissue Scaffolds , Transplantation, Heterologous
6.
Exp Dermatol ; 22(9): 601-3, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23947675

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is caused by deficiency of type VII collagen due to COL7A1 mutations such as c.6527insC, recurrently found in the Spanish RDEB population. Assessment of clonal correction-based therapeutic approaches for RDEB requires large expansions of cells, exceeding the replication capacity of human primary keratinocytes. Thus, immortalized RDEB cells with enhanced proliferative abilities would be valuable. Using either the SV40 large T antigen or papillomavirus HPV16-derived E6-E7 proteins, we immortalized and cloned RDEB keratinocytes carrying the c.6527insC mutation. Clones exhibited high proliferative and colony-forming features. Cytogenetic analysis revealed important differences between T antigen-driven and E6-E7-driven immortalization. Immortalized cells responded to differentiation stimuli and were competent for epidermal regeneration and recapitulation of the blistering RDEB phenotype in vivo. These features make these cell lines useful to test novel therapeutic approaches including those aimed at editing mutant COL7A1.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/therapy , Keratinocytes/metabolism , Mutation , Animals , Cell Line , Cell- and Tissue-Based Therapy , Epidermolysis Bullosa Dystrophica/pathology , Genetic Therapy , Heterografts , Homozygote , Humans , Keratinocytes/transplantation , Mice , Models, Genetic , Regeneration
7.
Mol Carcinog ; 51(4): 352-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21538579

ABSTRACT

Lysosomal cysteine protease cathepsin L (CTSL) is believed to play a role in tumor progression and is considered a marker for clinically invasive tumors. Studies from our laboratory using the classical mouse skin carcinogenesis model, with 7,12-dimethyl-benz[a]anthracene (DMBA) for initiation and 12-O-tetradecanoylphorbol-13-acetate (TPA) for promotion, showed that expression of CTSL is increased in papillomas and squamous cell carcinomas (SCC). We also carried out carcinogenesis studies using Ctsl-deficient nackt (nkt) mutant mice on three different inbred backgrounds. Unexpectedly, the multiplicity of papillomas was significantly higher in Ctsl-deficient than in wild-type mice on two unrelated backgrounds. Topical applications of TPA or DMBA alone to the skin of nkt/nkt mice did not induce papillomas, and there was no increase in spontaneous tumors in nkt/nkt mice on any of the three inbred backgrounds. Reduced epidermal cell proliferation in Ctsl-deficient nkt/nkt mice after TPA treatment suggested that they are not more sensitive than wild-type mice to TPA promotion. We also showed that deficiency of CTSL delays terminal differentiation of keratinocytes, and we propose that decreased elimination of initiated cells is at least partially responsible for the increased papilloma formation in the nackt model.


Subject(s)
Cathepsin L/physiology , Skin Neoplasms/prevention & control , Administration, Topical , Alleles , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Female , Genotype , Keratinocytes/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Knockout , Papilloma/metabolism , Polymorphism, Genetic , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tetradecanoylphorbol Acetate/administration & dosage
8.
Mol Carcinog ; 51(12): 973-83, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22006370

ABSTRACT

Studies show that elevated insulin-like growth factor-1 (IGF-1) levels are associated with an increased risk of breast cancer; however, mechanisms through which IGF-1 promotes mammary tumorigenesis in vivo have not been fully elucidated. To assess the possible involvement of COX-2 signaling in the pro-tumorigenic effects of IGF-1 in mammary glands, we used the unique BK5.IGF-1 mouse model in which transgenic (Tg) mice have significantly increased incidence of spontaneous and DMBA-induced mammary cancer compared to wild type (WT) littermates. Studies revealed that COX-2 expression was significantly increased in Tg mammary glands and tumors, compared to age-matched WTs. Consistent with this, PGE(2) levels were also increased in Tg mammary glands. Analysis of expression of the EP receptors that mediate the effects of PGE(2) showed that among the four G-protein-coupled receptors, EP3 expression was elevated in Tg glands. Up-regulation of the COX-2/PGE(2) /EP3 pathway was accompanied by increased expression of VEGF and a striking enhancement of angiogenesis in IGF-1 Tg mammary glands. Treatment with celecoxib, a selective COX-2 inhibitor, caused a 45% reduction in mammary PGE(2) levels, attenuated the influx of mast cells and reduced vascularization in Tg glands. These findings indicate that the COX-2/PGE(2) /EP3 signaling pathway is involved in IGF-1-stimulated mammary tumorigenesis and that COX-2-selective inhibitors may be useful in the prevention or treatment of breast cancer associated with elevated IGF-1 levels in humans. © 2011 Wiley Periodicals, Inc.


Subject(s)
Cyclooxygenase 2/metabolism , Insulin-Like Growth Factor I/physiology , Mammary Glands, Animal/enzymology , Signal Transduction/physiology , Animals , Celecoxib , Cyclooxygenase 2 Inhibitors/pharmacology , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Female , Mammary Glands, Animal/blood supply , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Neovascularization, Pathologic , Pyrazoles/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Prostaglandin E/metabolism , Sulfonamides/pharmacology
9.
Exp Dermatol ; 21(3): 217-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22379968

ABSTRACT

Strains of mice vary in their susceptibility to ultra-violet (UV) radiation-induced skin tumors. Some strains of hairless mice (homozygous for the spontaneous Hr(hr) mutation) are particularly susceptible to these tumors. The skin tumors that develop in hairless mice resemble, both at the morphologic and molecular levels, UV-induced squamous cell carcinomas (SCC) and their precursors in human. The most commonly employed hairless mice belong to the SKH1 stock. However, these mice are outbred and their genetic background is not characterized, which makes them a poor model for genetic studies. We have developed a new inbred strain from outbred SKH1 mice that we named SKHIN/Sprd (now at generation F31). In order to characterize the genetic background of this new strain, we genotyped a cohort of mice at F30 with 92 microsatellites and 140 single nucleotide polymorphisms (SNP) evenly distributed throughout the mouse genome. We also exposed SKHIN/Sprd mice to chronic UV irradiation and showed that they are as susceptible to UV-induced skin carcinogenesis as outbred SKH1 mice. In addition, we proved that, albeit with low efficiency, inbred SKHIN/Sprd mice are suitable for transgenic production by classical pronuclear microinjection. This new inbred strain will be useful for the development of transgenic and congenic strains on a hairless inbred background as well as the establishment of syngeneic tumor cell lines. These new tools can potentially help elucidate a number of features of the cutaneous response to UV irradiation in humans, including the effect of genetic background and modifier genes.


Subject(s)
Mice, Hairless/genetics , Models, Animal , Neoplasms, Radiation-Induced/genetics , Skin Neoplasms/genetics , Animals , Disease Models, Animal , Mice , Neoplasms, Radiation-Induced/etiology , Ultraviolet Rays
10.
Cells ; 11(16)2022 08 17.
Article in English | MEDLINE | ID: mdl-36010633

ABSTRACT

Diet is a critical environmental factor affecting breast cancer risk, and recent evidence shows that dietary exposures during early development can affect lifetime mammary cancer susceptibility. To elucidate the underlying mechanisms, we used our established crossover feeding mouse model, where exposure to a high-fat and high-sugar (HFHS) diet during defined developmental windows determines mammary tumor incidence and latency in carcinogen-treated mice. Mammary tumor incidence is significantly increased in mice receiving a HFHS post-weaning diet (high-tumor mice, HT) compared to those receiving a HFHS diet during gestation (low-tumor mice, LT). The current study revealed that the mammary stem cell (MaSC) population was significantly increased in mammary glands from HT compared to LT mice. Igf1 expression was increased in mammary stromal cells from HT mice, where it promoted MaSC self-renewal. The increased Igf1 expression was induced by DNA hypomethylation of the Igf1 Pr1 promoter, mediated by a decrease in Dnmt3b levels. Mammary tissues from HT mice also had reduced levels of Igfbp5, leading to increased bioavailability of tissue Igf1. This study provides novel insights into how early dietary exposures program mammary cancer risk, demonstrating that effective dietary intervention can reduce mammary cancer incidence.


Subject(s)
Dietary Exposure , Mammary Neoplasms, Animal , Animals , Carcinogens , Diet , Mammary Neoplasms, Animal/metabolism , Mice , Stem Cells/metabolism
11.
Breast Cancer Res Treat ; 130(2): 399-408, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21191649

ABSTRACT

Clinical studies show that estrogen receptor-α (ER) expressing tumors tend to have better prognosis, respond to antiestrogen therapy and have wild-type p53. Conversely, tumors with inactivating mutations in p53 tend to have worse outcomes and to be ER-negative and unresponsive to antihormone treatment. Previous studies from our laboratory have shown that p53 regulates ER expression transcriptionally, by binding the ER promoter and forming a complex with CARM1, CBP, c-Jun, RNA polymerase II and Sp1. In this study, the MMTV-Wnt-1 transgenic mouse model was used to demonstrate that p53 regulation of ER expression and function is not solely an in vitro phenomenon, but it is also operational in mammary tumorigenesis in vivo. The expression of ER and the ability to respond to tamoxifen were determined in mammary tumors arising in p53 wild type (WT) or p53 heterozygous (HT) animals carrying the Wnt-1 transgene. In p53 WT mice, development of ER-positive tumors was delayed by tamoxifen treatment, while tumors arising in p53 HT mice had significantly reduced levels of ER and were not affected by tamoxifen. P53 null tumors were also found in the p53 HT mice and these tumors were ER-negative. ER expression was upregulated in mouse mammary tumor cell lines following transfection with WT p53 or treatment with doxorubicin. These data demonstrate that p53 regulates ER expression in vivo, and affects response to tamoxifen. Results also provide an explanation for the concordant relationship between these prognostic proteins in human breast tumors.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/genetics , Tamoxifen/therapeutic use , Tumor Suppressor Protein p53/genetics , Animals , Cell Transformation, Neoplastic/genetics , Estrogen Receptor alpha/genetics , Female , Gene Expression , Genotype , Heterozygote , Loss of Heterozygosity , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Suppressor Protein p53/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
12.
Am J Pathol ; 177(4): 1958-68, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20724589

ABSTRACT

Citrullinemia type I (CTLN1, OMIM# 215700) is an inherited urea cycle disorder that is caused by an argininosuccinate synthetase (ASS) enzyme deficiency. In this report, we describe two spontaneous hypomorphic alleles of the mouse Ass1 gene that serve as an animal model of CTLN1. These two independent mouse mutant alleles, also described in patients affected with CTLN1, interact to produce a range of phenotypes. While some mutant mice died within the first week after birth, others survived but showed severe retardation during postnatal development as well as alopecia, lethargy, and ataxia. Notable pathological findings were similar to findings in human CTLN1 patients and included citrullinemia and hyperammonemia along with delayed cerebellar development, epidermal hyperkeratosis, and follicular dystrophy. Standard treatments for CTLN1 were effective in rescuing the phenotype of these mutant mice. Based on our studies, we propose that defective cerebellar granule cell migration secondary to disorganization of Bergmann glial cell fibers cause cerebellar developmental delay in the hyperammonemic and citrullinemic brain, pointing to a possible role for nitric oxide in these processes. These mouse mutations constitute a suitable model for both mechanistic and preclinical studies of CTLN1 and other hyperammonemic encephalopathies and, at the same time, underscore the importance of complementing knockout mutations with hypomorphic mutations for the generation of animal models of human genetic diseases.


Subject(s)
Argininosuccinate Synthase/physiology , Citrullinemia/etiology , Disease Models, Animal , Hyperammonemia/etiology , Mutation, Missense/genetics , Alleles , Animals , Arginine/pharmacology , Blotting, Western , Cell Movement , Cerebellum/abnormalities , Citrullinemia/drug therapy , Developmental Disabilities/drug therapy , Developmental Disabilities/etiology , Female , Growth Disorders/drug therapy , Growth Disorders/etiology , Humans , Hyperammonemia/drug therapy , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Nitric Oxide/metabolism , Phenotype , Sodium Benzoate/pharmacology , Syndrome
13.
Am J Pathol ; 174(5): 1869-79, 2009 May.
Article in English | MEDLINE | ID: mdl-19395652

ABSTRACT

The tumor suppressor phosphatase and tensin homolog (PTEN) is frequently involved in human prostate carcinoma. PTEN is therefore an attractive target for the development of preclinical animal models. Prostate intraepithelial neoplasia lesions develop in mice with Pten heterozygosity, but disease progression has been reported only in combination with either other tumor suppressor gene alterations or the conditional inactivation of both Pten alleles in prostate epithelial cells. We report that on a C57BL/6 background, in contrast to previous studies on mixed 129 genetic backgrounds, Pten locus heterozygosity is fully penetrant for the development of prostate adenocarcinoma. Grossly observable tumors were detected at 6 months of age, and, by 10 to 12 months, 100% of examined mice developed adenocarcinoma of the anterior prostate. Furthermore, double heterozygotes carrying both Pten and Tsc2-null alleles showed no increase relative to Pten(+/-) heterozygotes in either lesion development or progression. Lesions in both Pten(+/-); Tsc2(+/-), and Pten(+/-) mice exhibited loss of PTEN expression and activation of PI3K signaling. PI3K activation occurred early in prostate intraepithelial neoplasia lesion formation in these animals, consistent with loss of PTEN function, and contributed to the etiology of tumors that developed in Pten(+/-) mice. Furthermore, prostate lesion growth in Pten(+/-) mice was dependent on mTOR, as evidenced by a reduction in both phospho-S6 levels and proliferative index after rapamycin treatment.


Subject(s)
Adenocarcinoma/pathology , PTEN Phosphohydrolase/physiology , Prostatic Neoplasms/pathology , Protein Kinases/metabolism , Tumor Suppressor Proteins/physiology , Adenocarcinoma/metabolism , Animals , Blotting, Western , Disease Progression , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Incidence , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsatellite Repeats , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein
15.
Mol Carcinog ; 48(6): 508-16, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18942117

ABSTRACT

Cyclin D1b is an alternative transcript of the cyclin D1 gene (CCND1) expressed in human tumors. Its abundance is regulated by a single base pair polymorphism at the exon 4/intron 4 boundary (nucleotide 870). Epidemiological studies have shown a correlation between the presence of the G870A allele (that favors the splicing for cyclin D1b) with increased risk and less favorable outcome in several forms of cancer. More recently, it has been shown that, unlike cyclin D1a, the alternative transcript D1b by itself has the capacity to transform fibroblasts in vitro. In order to study the oncogenic potential of cyclin D1b, we developed transgenic mice expressing human cyclin D1b under the control of the bovine K5 promoter (K5D1b mice). Seven founders were obtained and none of them presented any significant phenotype or developed spontaneous tumors. Interestingly, K5D1b mice do not develop the fatal thymic hyperplasia, which is characteristic of the cyclin D1a transgenic mice (K5D1a). Susceptibility to skin carcinogenesis was tested in K5D1b mice using two-stage carcinogenesis protocols. In two independent experiments, K5D1b mice developed higher papilloma multiplicity as compared with wild-type littermates. However, when K5D1b mice were crossed with cyclin D1KO mice, the expression of cyclin D1b was unable to rescue the carcinogenesis-resistant phenotype of the cyclin D1 KO mice. To further explore the role of cyclin D1b in mouse models of carcinogenesis we carried out in silico analysis and in vitro experiments to evaluate the existence of a mouse homologous of the human cyclin D1b transcript. We were unable to find any evidence of an alternatively spliced transcript in mouse Ccnd1. These results show that human cyclin D1b has different biological functions than cyclin D1a and confirm its oncogenic properties.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin D1/genetics , Skin Neoplasms/genetics , Thymus Gland/pathology , Animals , Base Sequence , DNA Primers , Exons , Hyperplasia , Introns , Mice , Mice, Transgenic , Polymerase Chain Reaction , Polymorphism, Single Nucleotide
16.
Am J Pathol ; 173(2): 526-35, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18599613

ABSTRACT

It has been widely assumed that elevated CDK2 kinase activity plays a contributory role in tumorigenesis. We have previously shown that mice overexpressing CDK4 under control of the keratin 5 promoter (K5CDK4 mice) develop epidermal hyperplasia and increased susceptibility to squamous cell carcinomas. In this model, CDK4 overexpression results in increased CDK2 activity associated with the noncatalytic function of CDK4, sequestration of p21(Cip1) and p27(Kip1). Furthermore, we have shown that ablation of Cdk2 reduces Ras-Cdk4 tumorigenesis, suggesting that increased CDK2 activity plays an important role in Ras-mediated tumorigenesis. To investigate this hypothesis, we generated two transgenic mouse models of elevated CDK2 kinase activity, K5Cdk2 and K5Cdk4(D158N) mice. The D158N mutation blocks CDK4 kinase activity without interfering with its binding capability. CDK2 activation via overexpression of CDK4(D158N), but not of CDK2, resulted in epidermal hyperplasia. We observed elevated levels of p21(Cip1) in K5Cdk2, but not in K5Cdk4(D158N), epidermis, suggesting that CDK2 overexpression elicits a p21(Cip1) response to maintain keratinocyte homeostasis. Surprisingly, we found that neither CDK2 overexpression nor the indirect activation of CDK2 enhanced skin tumor development. Thus, although the indirect activation of CDK2 is sufficient to induce keratinocyte hyperproliferation, activation of CDK2 alone does not induce malignant progression in Ras-mediated tumorigenesis.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase 2/metabolism , Epidermis/enzymology , Keratinocytes/pathology , Skin Neoplasms/enzymology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Cell Transformation, Neoplastic/pathology , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Enzyme Activation , Epidermis/pathology , Keratinocytes/enzymology , Mice , Mice, Transgenic , Mutation , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology
17.
Am J Pathol ; 173(3): 824-34, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688034

ABSTRACT

Insulin-like growth factor-1 (IGF-1) stimulates proliferation, regulates tissue development, protects against apoptosis, and promotes the malignant phenotype in the breast and other organs. Some epidemiological studies have linked high circulating levels of IGF-1 with an increased risk of breast cancer. To study the role of IGF-1 in mammary tumorigenesis in vivo, we used transgenic mice in which overexpression of IGF-1 is under the control of the bovine keratin 5 (BK5) promoter and is directed to either the myoepithelial or basal cells in a variety of organs, including the mammary gland. This model closely recapitulates the paracrine exposure of breast epithelium to stromal IGF-1 seen in women. Histologically, mammary glands from transgenic mice were hyperplastic and highly vascularized. Mammary glands from prepubertal transgenic mice had significantly increased ductal proliferation compared with wild-type tissues, although this difference was not maintained after puberty. Transgenic mice also had increased susceptibility to mammary carcinogenesis, and 74% of the BK5.IGF-1 mice treated with 7,12-dimethylbenz[a]anthracene (20 microg/day) developed mammary tumors compared with 29% of the wild-type mice. Interestingly, 31% of the vehicle-treated BK5.IGF-1 animals, but none of the wild-type animals, spontaneously developed mammary cancer. The mammary tumors were moderately differentiated adenocarcinomas that expressed functional, nuclear estrogen receptor at both the protein and mRNA levels. These data support the hypothesis that tissue overexpression of IGF-1 stimulates mammary tumorigenesis.


Subject(s)
Adenocarcinoma/metabolism , Insulin-Like Growth Factor I/biosynthesis , Mammary Neoplasms, Experimental/metabolism , Paracrine Communication/physiology , Adenocarcinoma/pathology , Animals , Blotting, Western , Cattle , Cyclin D1/biosynthesis , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Keratin-5/biosynthesis , Keratin-8/biosynthesis , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Microscopy, Confocal , Promoter Regions, Genetic , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
18.
Toxicol Pathol ; 37(6): 733-40, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19679887

ABSTRACT

This study was performed to examine the carcinogenic effects of benzo[a]pyrene (B[a]P) and manufactured gas plant (MGP) residues on the hamster cheek pouch (HCP). Syrian hamsters were treated topically with a suspension of 2%, 10%, or 20% B[a]P or 50% or 100% MGP-7 (a mixture of residues from 7 MGP sites) in mineral oil for eight (short-term study) and sixteen, twenty, twenty-eight, and thirty-two weeks (long-term study). The short-term study showed that B[a]P induced p53 protein accumulation, indicative of genotoxic damage, as well as increased cell proliferation, hyperplasia, and inflammation, which is usually associated with promotional activity. In contrast, the MGP-7 presented only marginal p53 accumulation and induction of BrdU incorporation. In the long-term experiments, animals treated with 2% and 10% of B[a]P continued to show p53 protein accumulation as well as hyperplasia and increased cell proliferation and inflammation. By thirty weeks, all the animals treated with B[a]P had a 100% incidence of squamous cell carcinoma (SCC). Animals treated with 50% and 100% MGP-7 showed only weak hyperplasia and a low proliferation rate and accumulation of p53 protein through thirty-two weeks. Benzo[a]pyrene was highly carcinogenic when used at adequate doses. Manufactured gas plant residue, however, was not carcinogenic in this model.


Subject(s)
Benzo(a)pyrene/toxicity , Coal Tar/toxicity , Mouth Neoplasms/chemically induced , Animals , Carcinogenicity Tests/methods , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cheek/pathology , Cricetinae , Disease Models, Animal , Histocytochemistry , Industrial Waste , Male , Mesocricetus , Mouth Neoplasms/pathology , Research Design
19.
Oncogene ; 38(18): 3535-3550, 2019 05.
Article in English | MEDLINE | ID: mdl-30651598

ABSTRACT

Female breast cancer (BrCa) is the most common noncutaneous cancer among women in the United States. Human epidemiological studies reveal that a p53 single-nucleotide polymorphism (SNP) at codon 72, encoding proline (P72) or arginine (R72), is associated with differential risk of several cancers, including BrCa. However, the molecular mechanisms by which these variants affect mammary tumorigenesis remain unresolved. To investigate the effects of this polymorphism on susceptibility to mammary cancer, we used a humanized p53 mouse model, homozygous for either P72 or R72. Our studies revealed that R72 mice had a significantly higher mammary tumor incidence and reduced latency in both DMBA-induced and MMTV-Erbb2/Neu mouse mammary tumor models compared to P72 mice. Analyses showed that susceptible mammary glands from E-R72 (R72 x MMTV-Erbb2/Neu) mice developed a senescence-associated secretory phenotype (SASP) with influx of proinflammatory macrophages, ultimately resulting in chronic, protumorigenic inflammation. Mammary tumors arising in E-R72 mice also had an increased influx of tumor-associated macrophages, contributing to angiogenesis and elevated tumor growth rates. These results demonstrate that the p53 R72 variant increased susceptibility to mammary tumorigenesis through chronic inflammation.


Subject(s)
Carcinogenesis/genetics , Codon/genetics , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/pathology , Disease Models, Animal , Female , Humans , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Receptor, ErbB-2/genetics
20.
Orphanet J Rare Dis ; 14(1): 183, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31340837

ABSTRACT

BACKGROUND: Kindler Syndrome (KS) is a rare genodermatosis characterized by skin fragility, skin atrophy, premature aging and poikiloderma. It is caused by mutations in the FERMT1 gene, which encodes kindlin-1, a protein involved in integrin signalling and the formation of focal adhesions. Several reports have shown the presence of non-melanoma skin cancers in KS patients but a systematic study evaluating the risk of these tumors at different ages and their potential outcome has not yet been published. We have here addressed this condition in a retrospective study of 91 adult KS patients, characterizing frequency, metastatic potential and body distribution of squamous cell carcinoma (SCC) in these patients. SCC developed in 13 of the 91 patients. RESULTS: The youngest case arose in a 29-year-old patient; however, the cumulative risk of SCC increased to 66.7% in patients over 60 years of age. The highly aggressive nature of SCCs in KS was confirmed showing that 53.8% of the patients bearing SCCs develop metastatic disease. Our data also showed there are no specific mutations that correlate directly with the development of SCC; however, the mutational distribution along the gene appears to be different in patients bearing SCC from SCC-free patients. The body distribution of the tumor appearance was also unique and different from other bullous diseases, being concentrated in the hands and around the oral cavity, which are areas of high inflammation in this disease. CONCLUSIONS: This study characterizes SCCs in the largest series of KS patients reported so far, showing the high frequency and aggressiveness of these tumors. It also describes their particular body distribution and their relationship with mutations in the FERMT-1 gene. These data reinforce the need for close monitoring of premalignant or malignant lesions in KS patients.


Subject(s)
Blister/complications , Epidermolysis Bullosa/complications , Periodontal Diseases/complications , Photosensitivity Disorders/complications , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Adolescent , Adult , Aged , Blister/genetics , Epidermolysis Bullosa/genetics , Female , Humans , Male , Membrane Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , Periodontal Diseases/genetics , Photosensitivity Disorders/genetics , Skin Neoplasms/etiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL