Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Physiol ; 86: 453-478, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38345904

ABSTRACT

Studies in preclinical models support that the gut microbiota play a critical role in the development and progression of colorectal cancer (CRC). Specific microbial species and their corresponding virulence factors or associated small molecules can contribute to CRC development and progression either via direct effects on the neoplastic transformation of epithelial cells or through interactions with the host immune system. Induction of DNA damage, activation of Wnt/ß-catenin and NF-κB proinflammatory pathways, and alteration of the nutrient's availability and the metabolic activity of cancer cells are the main mechanisms by which the microbiota contribute to CRC. Within the tumor microenvironment, the gut microbiota alter the recruitment, activation, and function of various immune cells, such as T cells, macrophages, and dendritic cells. Additionally, the microbiota shape the function and composition of cancer-associated fibroblasts and extracellular matrix components, fashioning an immunosuppressive and pro-tumorigenic niche for CRC. Understanding the complex interplay between gut microbiota and tumorigenesis can provide therapeutic opportunities for the prevention and treatment of CRC.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Gastrointestinal Microbiome , Microbiota , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colonic Neoplasms/complications , Macrophages , Tumor Microenvironment
2.
Mol Cancer ; 23(1): 104, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755637

ABSTRACT

BACKGROUND: The faithful maintenance of DNA methylation homeostasis indispensably requires DNA methyltransferase 1 (DNMT1) in cancer progression. We previously identified DNMT1 as a potential candidate target for oral squamous cell carcinoma (OSCC). However, how the DNMT1- associated global DNA methylation is exploited to regulate OSCC remains unclear. METHODS: The shRNA-specific DNMT1 knockdown was employed to target DNMT1 on oral cancer cells in vitro, as was the use of DNMT1 inhibitors. A xenografted OSCC mouse model was established to determine the effect on tumor suppression. High-throughput microarrays of DNA methylation, bulk and single-cell RNA sequencing analysis, multiplex immunohistochemistry, functional sphere formation and protein immunoblotting were utilized to explore the molecular mechanism involved. Analysis of human samples revealed associations between DNMT1 expression, global DNA methylation and collaborative molecular signaling with oral malignant transformation. RESULTS: We investigated DNMT1 expression boosted steadily during oral malignant transformation in human samples, and its inhibition considerably minimized the tumorigenicity in vitro and in a xenografted OSCC model. DNMT1 overexpression was accompanied by the accumulation of cancer-specific DNA hypomethylation during oral carcinogenesis; conversely, DNMT1 knockdown caused atypically extensive genome-wide DNA hypomethylation in cancer cells and xenografted tumors. This novel DNMT1-remodeled DNA hypomethylation pattern hampered the dual activation of PI3K-AKT and CDK2-Rb and inactivated GSK3ß collaboratively. When treating OSCC mice, targeting DNMT1 achieved greater anticancer efficacy than the PI3K inhibitor, and reduced the toxicity of blood glucose changes caused by the PI3K inhibitor or combination of PI3K and CDK inhibitors as well as adverse insulin feedback. CONCLUSIONS: Targeting DNMT1 remodels a novel global DNA hypomethylation pattern to facilitate anticancer efficacy and minimize potential toxic effects via balanced signaling synergia. Our study suggests DNMT1 is a crucial gatekeeper regarding OSCC destiny and treatment outcome.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Gene Expression Regulation, Neoplastic , Mouth Neoplasms , Humans , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Animals , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mice , Cell Line, Tumor , Xenograft Model Antitumor Assays , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Signal Transduction , Cell Proliferation
3.
Clin Immunol ; 263: 110203, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575046

ABSTRACT

Langerhans cell histiocytosis (LCH) is characterized by an expansion and accumulation of pathological histiocytes expressing langerin (CD207) and CD1a in different organs under an inflammatory milieu. The origin of pathognomonic precursors of LCH is widely debated, but monocytes and pre-dendritic cells (pre-DC) play a significant role. Remarkably, we found an expansion of AXLhigh cells in the CD11c+ subset of patients with active LCH, which also express the pathognomonic CD207 and CD1a. Moreover, we obtained a monocyte-derived LC-like (mo-LC-like) expressing high levels of AXL when treated with inflammatory cytokine, or plasma of patients with active disease. Intriguingly, inhibiting the mTOR pathway at the initial stages of monocyte differentiation to LC-like fosters the pathognomonic LCH program, highly increasing CD207 levels, together with NOTCH1 induction. We define here that AXLhigh could also be taken as a strong pathognomonic marker for LCH, and the release of Langerin and NOTCH1 expression depends on the inhibition of the mTOR pathway.


Subject(s)
Antigens, CD , Axl Receptor Tyrosine Kinase , Histiocytosis, Langerhans-Cell , Lectins, C-Type , Mannose-Binding Lectins , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , TOR Serine-Threonine Kinases , Female , Humans , Male , Antigens, CD/metabolism , Antigens, CD1/metabolism , Biomarkers , Cell Differentiation , Histiocytosis, Langerhans-Cell/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Monocytes/metabolism , Monocytes/immunology , Myeloid Cells/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Notch1/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
4.
Int J Mol Sci ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38279352

ABSTRACT

Specifying the role of genetic mutations in cancer development is crucial for effective screening or targeted treatments for people with hereditary cancer predispositions. Our goal here is to find the relationship between a number of cancerogenic mutations and the probability of cancer induction over the lifetime of cancer patients. We believe that the Avrami-Dobrzynski biophysical model can be used to describe this mechanism. Therefore, clinical data from breast and ovarian cancer patients were used to validate this model of cancer induction, which is based on a purely physical concept of the phase-transition process with an analogy to the neoplastic transformation. The obtained values of model parameters established using clinical data confirm the hypothesis that the carcinogenic process strongly follows fractal dynamics. We found that the model's theoretical prediction and population clinical data slightly differed for patients with the age below 30 years old, and that might point to the existence of an ancillary protection mechanism against cancer development. Additionally, we reveal that the existing clinical data predict breast or ovarian cancers onset two years earlier for patients with BRCA1/2 mutations.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Humans , Female , Adult , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/epidemiology , Mutation , Genetic Predisposition to Disease , Breast Neoplasms/genetics
5.
Tumour Biol ; 45(1): 95-110, 2023.
Article in English | MEDLINE | ID: mdl-37742670

ABSTRACT

BACKGROUND: Epithelial-mesenchymal transition (EMT) is associated with altered cellular adhesion. We previously demonstrated that cellular adhesion influences Low-dose Hyper-Radiosensitivity (HRS) in a variety of tumor cells. However, the relationship of low-dose HRS with the phenotypic plasticity incurred by EMT during the neoplastic transformation remains to be elucidated. OBJECTIVE: To investigate whether acquisition of EMT phenotype during progressive neoplastic transformation may affect low-dose radiation sensitivity. METHODS: Primary thyroid cells obtained from a human cystic thyroid nodule were first subjected to nutritional stress. This yielded immortalized INM-Thy1 cell strain, which was further treated with either multiple γ-radiation fractions (1.5 Gy each) or repetitive cycles of 3-methylcholanthrene and phorbol-12-myristate-13-acetate, yielding two progressive transformants, viz., INM-Thy1R and INM-Thy1C. Morphological alterations, chromosomal double-minutes, cell adhesion proteins, anchorage dependency, tumorigenicity in nude mice and cellular radiosensitivity were studied in these strains. RESULTS: Both transformants (INM-Thy1R, INM-Thy1C) displayed progressive tumorigenic features, viz., soft agar colony growth and solid tumor growth in nude mice, coupled with features of epithelial-mesenchymal transition and activated Wnt pathway. Incidentally, the chemical-induced transformant (INM-Thy1C) displayed a prominent HRS (αs/αr = 29.35) which remained unaffected at high cell density. However, the parental (INM-Thy1) cell line as well as radiation-induced transformant (INM-Thy1R) failed to show this hypersensitivity. CONCLUSION: The study shows that induction of EMT in thyroid follicular cells may accompany increased susceptibility to low-dose ionizing radiation, which was attenuated by adaptive resistance acquired during radiation-induced transformation.


Subject(s)
Epithelial-Mesenchymal Transition , Thyroid Epithelial Cells , Animals , Mice , Humans , Cell Adhesion/genetics , Epithelial-Mesenchymal Transition/genetics , Mice, Nude , Carcinogenesis
6.
Appl Microbiol Biotechnol ; 107(7-8): 2097-2109, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36881118

ABSTRACT

The biological function of the agrobacterial oncogene rolA is very poorly understood compared to other components of the mechanism of horizontal gene transfer during agrobacterial colonization of plants. Research groups around the world have worked on this problem, and available information is reviewed in this review, but other rol oncogenes have been studied much more thoroughly. Having one unexplored element makes it impossible to form a complete picture. However, the limited data suggest that the rolA oncogene and its regulatory apparatus have great potential in plant biotechnology and genetic engineering. Here, we collect and discuss available experimental data about the function and structure of rolA. There is still no clear understanding of the mechanism of RolA and its structure and localization. We believe this is because of the nucleotide structure of a frameshift in the most well-studied rolA gene of the agropine type pRi. In fact, interest in the genes of agrobacteria as natural tools for the phenotypic or biochemical engineering of plants increased. We believe that a detailed understanding of the molecular mechanisms will be forthcoming. KEY POINTS: • Among pRi T-DNA oncogenes, rolA is the least understood in spite of many studies. • Frameshift may be the reason for the failure to elucidate the role of agropine rolA. • Understanding of rolA is promising for the phenotypic and biochemical engineering of plants.


Subject(s)
Gene Transfer, Horizontal , Rhizobium , Plants, Genetically Modified , DNA , Genetic Engineering , Oncogenes , Rhizobium/genetics
7.
Planta ; 256(1): 8, 2022 Jun 11.
Article in English | MEDLINE | ID: mdl-35690636

ABSTRACT

MAIN CONCLUSION: Increased flavonol accumulation and enhanced drought tolerance in A4-rolB-overexpressing plants can be explained by the cooperative action of the SA and ROS signalling pathways. Clarification of function of the A4-rolB plast gene from pRiA4 of Rhizobium rhizogenes will allow a better understanding of the biological principles of the natural transformation process and its use as a tool for plant bioengineering. In the present study, we investigated whether the overexpression of A4-rolB gene could regulate two important processes, flavonoid biosynthesis and drought tolerance. In addition, we investigated some aspects of the possible machinery of the A4-rolB-induced changes in plant physiology, such as crosstalk of the major signalling systems. Based on the data obtained in this work, it can be presumed that constitutive overexpression of A4-rolB leads to the activation of the salicylic acid signalling system. An increase in flavonol accumulation and enhanced drought tolerance can be explained by the cooperative action of SA and ROS pathways.


Subject(s)
Arabidopsis , Agrobacterium , Arabidopsis/genetics , Droughts , Flavonoids/metabolism , Flavonols/metabolism , Homeostasis , Hormones/metabolism , Plants, Genetically Modified/metabolism , Reactive Oxygen Species/metabolism
8.
Radiat Environ Biophys ; 61(1): 169-175, 2022 03.
Article in English | MEDLINE | ID: mdl-34665303

ABSTRACT

The nucleation and growth theory, described by the Avrami equation (also called Johnson-Mehl-Avrami-Kolmogorov equation), and usually used to describe crystallization and nucleation processes in condensed matter physics, was applied in the present paper to cancer physics. This can enhance the popular multi-hit model of carcinogenesis to volumetric processes of single cell's DNA neoplastic transformation. The presented approach assumes the transforming system as a DNA chain including many oncogenic mutations. Finally, the probability function of the cell's cancer transformation is directly related to the number of oncogenic mutations. This creates a universal sigmoidal probability function of cancer transformation of single cells, as observed in the kinetics of nucleation and growth, a special case of a phase transition process. The proposed model, which represents a different view on the multi-hit carcinogenesis approach, is tested on clinical data concerning gastric cancer. The results also show that cancer transformation follows DNA fractal geometry.


Subject(s)
DNA , Neoplasms , Carcinogenesis/genetics , Crystallization , Humans , Kinetics
9.
Int J Mol Sci ; 23(4)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35216085

ABSTRACT

The equine sarcoid is one of the most common neoplasias in the Equidae family. Despite the association of this tumor with the presence of bovine papillomavirus (BPV), the molecular mechanism of this lesion has not been fully understood. The transgenization of equine adult cutaneous fibroblast cells (ACFCs) was accomplished by nucleofection, followed by detection of molecular modifications using high-throughput NGS transcriptome sequencing. The results of the present study confirm that BPV-E4- and BPV-E1^E4-mediated nucleofection strategy significantly affected the transcriptomic alterations, leading to sarcoid-like neoplastic transformation of equine ACFCs. Furthermore, the results of the current investigation might contribute to the creation of in vitro biomedical models suitable for estimating the fates of molecular dedifferentiability and the epigenomic reprogrammability of BPV-E4 and BPV-E4^E1 transgenic equine ACFC-derived sarcoid-like cell nuclei in equine somatic cell-cloned embryos. Additionally, these in vitro models seem to be reliable for thoroughly recognizing molecular mechanisms that underlie not only oncogenic alterations in transcriptomic signatures, but also the etiopathogenesis of epidermal and dermal sarcoid-dependent neoplastic transformations in horses and other equids. For those reasons, the aforementioned transgenic models might be useful for devising clinical treatments in horses afflicted with sarcoid-related neoplasia of cutaneous and subcutaneous tissues.


Subject(s)
Fibroblasts/virology , Horse Diseases/virology , Horses/virology , Neoplasms/virology , Papillomaviridae/genetics , Sarcoidosis/virology , Skin Diseases/virology , Animals , Animals, Genetically Modified/virology , Equidae/virology , Papillomavirus Infections/virology , Skin/virology , Transcriptome/genetics
10.
Ecotoxicol Environ Saf ; 208: 111752, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33396077

ABSTRACT

Arsenic is a toxic heavy metal vastly dispersed all over the earth crust. It manifests several major adverse health issues to millions of arsenic exposed populations. Arsenic is associated with different types of cancer, cardiovascular disorders, diabetes, hypertension and many other diseases. On the contrary, arsenic (arsenic trioxide, As2O3) is used as a chemotherapeutic agent in the treatment of acute promyelocytic leukemia. Balance between arsenic induced cellular proliferations and apoptosis finally decide the outcome of its transformation rate. Arsenic propagates signals via cellular and nuclear pathways depending upon the chemical nature, and metabolic-fates of the arsenical compounds. Arsenic toxicity is propagated via ROS induced stress to DNA-repair mechanism and mitochondrial stability in the cell. ROS induced alteration in p53 regulation and some mitogen/ oncogenic functions determine the transformation outcome influencing cyclin-cdk complexes. Growth factor regulator proteins such as c-Jun, c-fos and c-myc are influenced by chronic arsenic exposure. In this review we have delineated arsenic induced ROS regulations of epidermal growth factor receptor (EGFR), NF-ĸß, MAP kinase, matrix-metalloproteinases (MMPs). The role of these signaling molecules has been discussed in relation to cellular apoptosis, cellular proliferation and neoplastic transformation. The arsenic stimulated pathways which help in proliferation and neoplastic transformation ultimately resulted in cancer manifestation whereas apoptotic pathways inhibited carcinogenesis. Therapeutic strategies against arsenic should be designed taking into account all these factors.


Subject(s)
Arsenic/physiology , Cell Proliferation/physiology , Plastics/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenic/metabolism , Arsenic Trioxide/metabolism , Arsenic Trioxide/pharmacology , Arsenicals/metabolism , Cell Proliferation/drug effects , Humans , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasms , Oxides/toxicity , Signal Transduction/drug effects
11.
Int J Mol Sci ; 22(21)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34769230

ABSTRACT

Nandrolone (Ndn) and boldenone (Bdn), the synthetic testosterone analogues with strong anabolic effects, despite being recognized as potentially carcinogenic compounds, are commonly abused by athletes and bodybuilders, which includes women, worldwide. This study tested the hypothesis that different doses of Ndn and Bdn can initiate neoplastic transformation of porcine ovarian putative stem cells (poPSCs). Immunomagnetically isolated poPSCs were expanded ex vivo in the presence of Ndn or Bdn, for 7 and 14 days. Results show that pharmacological doses of both Ndn and Bdn, already after 7 days of poPSCs culture, caused a significant increase of selected, stemness-related markers of cancer cells: CD44 and CD133. Notably, Ndn also negatively affected poPSCs growth not only by suppressing their proliferation and mitochondrial respiration but also by inducing apoptosis. This observation shows, for the first time, that chronic exposure to Ndn or Bdn represents a precondition that might enhance risk of poPSCs neoplastic transformation. These studies carried out to accomplish detailed molecular characterization of the ex vivo expanded poPSCs and their potentially cancerous derivatives (PCDs) might be helpful to determine their suitability as nuclear donor cells (NDCs) for further investigations focused on cloning by somatic cell nuclear transfer (SCNT). Such investigations might also be indispensable to estimate the capabilities of nuclear genomes inherited from poPSCs and their PCDs to be epigenetically reprogrammed (dedifferentiated) in cloned pig embryos generated by SCNT. This might open up new possibilities for biomedical research aimed at more comprehensively recognizing genetic and epigenetic mechanisms underlying not only tumorigenesis but also reversal/retardation of pro-tumorigenic intracellular events.


Subject(s)
Cell Transformation, Neoplastic , Cellular Reprogramming/drug effects , Nandrolone/adverse effects , Ovarian Neoplasms , Ovary , Stem Cells , Testosterone/analogs & derivatives , Animals , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Nandrolone/pharmacology , Ovarian Neoplasms/chemically induced , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Stem Cells/metabolism , Stem Cells/pathology , Swine , Testosterone/adverse effects , Testosterone/pharmacology
12.
Mol Carcinog ; 57(1): 114-124, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28926134

ABSTRACT

Chromosome instability (CIN) is widely observed in both sporadic and hereditary colorectal cancer (CRC). Defects in APC and WNT signaling are primarily associated with CIN in hereditary CRC, but the genetic causes for CIN in sporadic CRC remain elusive. Using high-density SNP array and exome data from The Cancer Genome Atlas (TCGA), we characterized loss of heterozygosity (LOH) and copy number variation (CNV) in the peripheral blood, normal colon, and corresponding tumor tissue in 15 CRC patients with proficient mismatch repair (MMR) and 24 CRC patients with deficient MMR. We found a high frequency of 18q LOH in tumors and arm-specific enrichment of genetic aberrations on 18q in the normal colon (primarily copy neutral LOH) and blood (primarily copy gain). These aberrations were specific to the sporadic, pMMR CRC. Though in tumor samples genetic aberrations were observed for genes commonly mutated in hereditary CRC (eg, APC, CTNNB1, SMAD4, BRAF), none of them showed LOH or CNV in the normal colon or blood. DCC located on 18q21.1 topped the list of genes with genetic aberrations in the tumor. In an independent cohort of 13 patients subjected to Whole Genome Sequencing (WGS), we found LOH and CNV on 18q in adenomatous polyp and tumor tissues. Our data suggests that patients with sporadic CRC may have genetic aberrations preferentially enriched on 18q in their blood, normal colon epithelium, and non-malignant polyp lesions that may prove useful as a clinical marker for sporadic CRC detection and risk assessment.


Subject(s)
Colorectal Neoplasms/genetics , DNA Copy Number Variations , DNA Mismatch Repair/genetics , Loss of Heterozygosity , Aged , Aged, 80 and over , Chromosomal Instability , Chromosomes, Human, Pair 18/genetics , Cohort Studies , Colorectal Neoplasms/pathology , Female , Genotype , Humans , Male , Middle Aged , Mutation
13.
Ann Hematol ; 97(1): 17-29, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29043381

ABSTRACT

Follicular Lymphoma (FL) is the second most common type of non-Hodgkin lymphoma and is considered to be the prototype of indolent lymphomas. Histologic transformation into an aggressive lymphoma, which is expected to occur at a rate of 2 to 3% each year, is associated with rapid progression, treatment resistance, and poor prognosis. Recent modifications to the physiopathologic mechanism of transformed follicular lymphoma (t-FL) have been proposed, including genetic and epigenetic mechanisms as well as a role for the microenvironment. Although t-FL is considered a devastating complication, as it is associated with treatment-refractory disease and a dismal outcome, recent data in the rituximab era have suggested that not only is the prognosis less severe than reported in the previous literature but the risk of transformation is also lower. Thus, this study aimed to review the most recent research on t-FL in an attempt to better understand the clinical meaning of transformation from FL to diffuse large B cell lymphoma (DLBCL) and the impact of current treatment strategies on the curability of this intriguing subentity of lymphoma.


Subject(s)
Cell Transformation, Neoplastic , Lymphoma, Follicular/pathology , Cell Transformation, Neoplastic/genetics , Disease Progression , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Recurrence, Local , Treatment Outcome
14.
Int J Mol Sci ; 19(12)2018 Dec 04.
Article in English | MEDLINE | ID: mdl-30518128

ABSTRACT

The cholangipathies are a class of liver diseases that specifically affects the biliary tree. These pathologies may have different etiologies (genetic, autoimmune, viral, or toxic) but all of them are characterized by a stark inflammatory infiltrate, increasing overtime, accompanied by an excess of periportal fibrosis. The cellular types that mount the regenerative/reparative hepatic response to the damage belong to different lineages, including cholagiocytes, mesenchymal and inflammatory cells, which dynamically interact with each other, exchanging different signals acting in autocrine and paracrine fashion. Those messengers may be proinflammatory cytokines and profibrotic chemokines (IL-1, and 6; CXCL1, 10 and 12, or MCP-1), morphogens (Notch, Hedgehog, and WNT/ß-catenin signal pathways) and finally growth factors (VEGF, PDGF, and TGFß, among others). In this review we will focus on the main molecular mechanisms mediating the establishment of a fibroinflammatory liver response that, if perpetuated, can lead not only to organ dysfunction but also to neoplastic transformation. Primary Sclerosing Cholangitis and Congenital Hepatic Fibrosis/Caroli's disease, two chronic cholangiopathies, known to be prodrome of cholangiocarcinoma, for which several murine models are also available, were also used to further dissect the mechanisms of fibroinflammation leading to tumor development.


Subject(s)
Bile Duct Diseases/pathology , Liver Cirrhosis/pathology , Liver/injuries , Precancerous Conditions/pathology , Animals , Epithelium/pathology , Humans , Liver/pathology , Mesoderm/pathology
15.
Mol Carcinog ; 55(2): 161-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25594762

ABSTRACT

The functional role of nuclear factor of activated T-cells (NFAT), while it has been extensively investigated in the immune system, remains uncertain in bladder cancer development. We here aim to assess the effects of cyclosporine A (CsA) and tacrolimus (FK506), immunosuppressants known to specifically inactivate the NFAT pathway in immune cells, on neoplastic transformation of urothelial cells. Immunohistochemistry revealed that the expression levels of NFATc1, a NFAT isoform shown to function as an oncogene in a sarcoma model, were elevated in urothelial neoplasms, compared with non-neoplastic urothelial tissues, and in low-grade and high-grade papillary urothelial carcinomas, compared with papillary urothelial neoplasms of low malignant potential. In an immortalized normal urothelial cell line SVHUC, CsA and FK506 reduced NFATc1 expression, NFAT transcriptional activity, and the expression of c-myc, a downstream target of NFATc1 signals. Treatment with CsA or FK506 in the SVHUC cells undergoing neoplastic transformation induced by exposure to a chemical carcinogen 3-methylcholanthrene resulted in strong inhibition in colony formation in vitro as well as tumor formation in NOD-SCID mice. CsA and FK506 were additionally found to up-regulate the expression of several molecules that play a protective role in bladder tumorigenesis, including p53, p21, and p27, and down-regulate that of oncogenic genes, such as cyclin D1, cyclin D3, and cyclin E, in SVHUC cells with the carcinogen challenge. Thus, CsA and FK506 likely inhibit urothelial tumorigenesis. These findings offer a potential chemopreventive approach for urothelial tumors using NFAT inhibitors.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cyclosporine/administration & dosage , NFATC Transcription Factors/metabolism , Tacrolimus/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Animals , Cell Line , Cyclosporine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Methylcholanthrene/adverse effects , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/drug effects , Tacrolimus/pharmacology , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Chromosome Res ; 23(4): 663-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25957863

ABSTRACT

The molecular mechanisms underlying spontaneous neoplastic transformation in cultured mammalian cells remain poorly understood, confounding recognition of parallels with the biology of naturally occurring cancer. The broad use of tumorigenic canine cell lines as research tools, coupled with the accumulation of cytogenomic data from naturally occurring canine cancers, makes the domestic dog an ideal system in which to investigate these relationships. We developed a canine kidney cell line, CKB1-3T7, which allows prospective examination of the onset of spontaneous immortalization and tumorigenicity. We documented the accumulation of cytogenomic aberrations in CKB1-3T7 over 24 months in continuous culture. The majority of aberrations emerged in parallel with key phenotypic changes in cell morphology, growth kinetics, and tumor incidence and latency. Focal deletion of CDKN2A/B emerged first, preceding the onset and progression of tumorigenic potential, and progressed to a homozygous deletion across the cell population during extended culture. Interestingly, CKB1-3T7 demonstrated a tumorigenic phenotype in vivo prior to exhibiting loss of contact inhibition in vitro. We also performed the first genome-wide characterization of the canine tumorigenic cell line MDCK, which also exhibited CDKN2A/B deletion. MDCK and CKB1-3T7 cells shared several additional aberrations that we have reported previously as being highly recurrent in spontaneous canine cancers, many of which, as with CDKN2A/B deletion, are evolutionarily conserved in their human counterparts. The conservation of these molecular events across multiple species, in vitro and in vivo, despite their contrasting karyotypic architecture, is a powerful indicator of a common mechanism underlying emerging neoplastic activity. Through integrated cytogenomic and phenotypic characterization of serial passages of CKB1-3T7 from initiation to development of a tumorigenic phenotype, we present a robust and readily accessible model (to be made available through the American Type Culture Collection) of spontaneous neoplastic transformation that overcomes many of the limitations of earlier studies.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosome Aberrations , Karyotype , Neoplasms/genetics , Animals , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cells, Cultured , DNA Copy Number Variations , Dogs , In Situ Hybridization, Fluorescence , Madin Darby Canine Kidney Cells , Male , Neoplasms/pathology
17.
Curr Genomics ; 17(1): 33-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27013923

ABSTRACT

Research on bladder neoplasms in pediatric and teen patients (BNPTP) has described 21 genes, which are variously involved in this disease and are mostly responsible for deregulated cell proliferation. However, due to the limited number of publications on this subject, it is still unclear what type of relationships there are among these genes and which are the chances that, while having different molecular functions, they i) act as downstream effector genes of well-known pro- or anti- proliferative stimuli and/or interplay with biochemical pathways having oncological relevance or ii) are specific and, possibly, early biomarkers of these pathologies. A Gene Ontology (GO)-based analysis showed that these 21 genes are involved in biological processes, which can be split into two main classes: cell regulation-based and differentiation/development-based. In order to understand the involvement/overlapping with main cancer-related pathways, we performed a meta-analysis dependent on the 189 oncogenic signatures of the Molecular Signatures Database (OSMSD) curated by the Broad Institute. We generated a binary matrix with 53 gene signatures having at least one hit; this analysis i) suggests that some genes of the original list show inconsistencies and might need to be experimentally re- assessed or evaluated as biomarkers (in particular, ACTA2) and ii) allows hypothesizing that important (proto)oncogenes (E2F3, ERBB2/HER2, CCND1, WNT1, and YAP1) and (putative) tumor suppressors (BRCA1, RBBP8/CTIP, and RB1-RBL2/p130) may participate in the onset of this disease or worsen the observed phenotype, thus expanding the list of possible molecular targets for the treatment of BNPTP.

18.
Mar Drugs ; 14(7)2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27428983

ABSTRACT

Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1-6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1-4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P⁺ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.


Subject(s)
Alkaloids/pharmacology , Cell Transformation, Neoplastic/drug effects , Epidermal Growth Factor/metabolism , Guanidines/pharmacology , Porifera/chemistry , Alkaloids/chemistry , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Guanidine/analogs & derivatives , Guanidine/chemistry , Guanidine/pharmacology , Guanidines/chemistry , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Proc Natl Acad Sci U S A ; 110(34): 13927-31, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23908399

ABSTRACT

Individuals suffering from type 2 diabetes or obesity exhibit a significant increase in the incidence of various types of cancer. It is generally accepted that those conditions arise from overnutrition and a sedentary lifestyle, which lead to insulin resistance characterized by overproduction of insulin acting as a growth factor. There is a consensus based largely on epidemiological data that chronic overproduction of insulin is responsible for the increased incidence of cancer. A model system in culture of NIH 3T3 cells induces the collective effects of serum growth factors on progression through the stages of field cancerization. It shows that the driving force of progression is promotion of cell growth under selection at high cell density, with no requirement for exogenous carcinogenic agents. The early effect is gradual selection among many preexisting, low-penetrance preneoplastic mutations or stable epigenetic variants, followed by sporadic, high-penetrance transforming variants, all dependent on endogenous processes. The significance of the results for cancer in diabetic and obese individuals is that the initial stages of the process involve multiorgan metabolic interactions that produce a systemic insulin resistance with chronic overproduction of insulin and localized field cancerization. Hypomagnesemia is prevalent in the foregoing metabalo/systemic disorders, and may also provide a selective microenvironment for tumor development.


Subject(s)
Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/complications , Hyperinsulinism/complications , Intercellular Signaling Peptides and Proteins/blood , Models, Biological , Neoplasms/etiology , Obesity/complications , Animals , Hyperinsulinism/etiology , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , NIH 3T3 Cells
20.
Vet Radiol Ultrasound ; 56(3): 257-63, 2015.
Article in English | MEDLINE | ID: mdl-25393217

ABSTRACT

Neoplastic transformation of Spirocerca lupi induced esophageal nodules carries a poor prognosis. Clinical, clinicopathological, endoscopic, and radiographic characteristics may be indicative of neoplastic transformation but variable sensitivity and specificity of these parameters makes their use questionable. We hypothesized that CT would be a better diagnostic modality to discriminate between non-neoplastic and neoplastic nodules. In this prospective study of 38 dogs, the appearance and perfusion characteristics of confirmed spirocercosis-induced neoplastic and non-neoplastic esophageal nodules were described using survey CT and triple phase dynamic CT angiography (CTA). Pre- and post-contrast early arterial, late arterial, and venous CTA images were evaluated. Non-neoplastic nodules were smooth and nonmineralized with a higher proportion of hypoattenuating necropurulent cavities compared to neoplastic nodules that had a more irregular surface, with 93% having mineralized foci and rarely any hypoattenuating pockets. Non-neoplastic nodules were significantly more perfused than neoplastic nodules with the difference being up to 23 Hounsfield units. The difference was most marked in the early and late arterial phases (P = 0.0005 and 0.00005, respectively). Ratios of the normal esophagus adjacent to the neoplastic and non-neoplastic nodules did not differ significantly from each other. Perfusion findings demonstrated relative hypoperfusion of the esophageal sarcomas. Findings from the current study indicated that CT characteristics of relative postcontrast hypoperfusion, combined with nodule irregularity and mineralization warrant a high level of concern for neoplastic transformation in canine spirocercosis-induced esophageal nodules.


Subject(s)
Dog Diseases/diagnostic imaging , Esophageal Neoplasms/veterinary , Sarcoma/veterinary , Spirurida Infections/veterinary , Thelazioidea/physiology , Animals , Dog Diseases/parasitology , Dogs , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/parasitology , Female , Male , Prospective Studies , Sarcoma/diagnostic imaging , Sarcoma/parasitology , Spirurida Infections/diagnostic imaging , Spirurida Infections/parasitology , Tomography, X-Ray Computed/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL