35 results on '"Marcella Sini"'
Search Results
2. Supplementary Table 1 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Table 1 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 5 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Figure 5 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure 3 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Figure 3 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure 4 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Figure 4 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table 3 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Table 3 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table 2 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Table 2 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Sustained activation of extracellular signal-regulated kinase (ERK) has been detected previously in numerous tumors in the absence of RAS-activating mutations. However, the molecular mechanisms responsible for ERK-unrestrained activity independent of RAS mutations remain unknown. Here, we evaluated the effects of the functional interactions of ERK proteins with dual-specificity phosphatase 1 (DUSP1), a specific inhibitor of ERK, and S-phase kinase-associated protein 2 (SKP2)/CDC28 protein kinase 1b (CKS1) ubiquitin ligase complex in human hepatocellular carcinoma (HCC). Levels of DUSP1, as assessed by real-time reverse transcription–PCR and Western blot analysis, were significantly higher in tumors with better prognosis (as defined by the length of patients' survival) when compared with both normal and nontumorous surrounding livers, whereas DUSP1 protein expression sharply declined in all HCC with poorer prognosis. In the latter HCC subtype, DUSP1 inactivation was due to either ERK/SKP2/CKS1-dependent ubiquitination or promoter hypermethylation associated with loss of heterozygosity at the DUSP1 locus. Noticeably, expression levels of DUSP1 inversely correlated with those of activated ERK, as well as with proliferation index and microvessel density, and directly with apoptosis and survival rate. Subsequent functional studies revealed that DUSP1 reactivation led to suppression of ERK, CKS1, and SKP2 activity, inhibition of proliferation and induction of apoptosis in human hepatoma cell lines. Taken together, the present data indicate that ERK achieves unrestrained activity during HCC progression by triggering ubiquitin-mediated proteolysis of its specific inhibitor DUSP1. Thus, DUSP1 may represent a valuable prognostic marker and ERK, CKS1, or SKP2 potential therapeutic targets for human HCC. [Cancer Res 2008;68(11):4192–200]
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 2 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Figure 2 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 1 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Rosa M. Pascale, Francesco Feo, Maria R. Muroni, Maria A. Seddaiu, Maria L. Tomasi, Maddalena Frau, Patrizia Virdis, Maria R. De Miglio, Maria M. Simile, Lucia Daino, Marcella Sini, Rossella Pellegrino, Sara Ladu, Floriana Meloni, Federico Pinna, and Diego F. Calvisi
- Abstract
Supplementary Figure 1 from Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Published
- 2023
- Full Text
- View/download PDF
11. Universal toxin-based selection for precise genome engineering in human cells
- Author
-
Mohammad Bohlooly-Y, Nina Akrap, Benjamin J. M. Taylor, Euan Gordon, Grzegorz Sienski, Mike Firth, Giovanni Ciotta, Anders Lundin, Matthew A. Coelho, Silvia Cerboni, Aleksandra Sieńska, Marcella Sini, Marcello Maresca, Luna Simona Pane, Giovanni Pellegrini, Xiufeng Xu, Songyuan Li, Suman Mitra, Bojana Lazovic, Sandra Wimberger, Carl Möller, Michelle J. Porritt, University of Zurich, Li, Songyuan, Sienski, Grzegorz, and Maresca, Marcello
- Subjects
0301 basic medicine ,Science ,Population ,10184 Institute of Veterinary Pathology ,General Physics and Astronomy ,1600 General Chemistry ,Genetics and Molecular Biology ,Computational biology ,Gene delivery ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Genome engineering ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genome editing ,1300 General Biochemistry, Genetics and Molecular Biology ,Recombinase ,education ,Gene ,education.field_of_study ,Multidisciplinary ,Biological techniques ,General Chemistry ,3100 General Physics and Astronomy ,030104 developmental biology ,chemistry ,General Biochemistry ,Genetic engineering ,570 Life sciences ,biology ,Stem cell ,Genetic techniques ,030217 neurology & neurosurgery ,DNA ,Biotechnology - Abstract
Prokaryotic restriction enzymes, recombinases and Cas proteins are powerful DNA engineering and genome editing tools. However, in many primary cell types, the efficiency of genome editing remains low, impeding the development of gene- and cell-based therapeutic applications. A safe strategy for robust and efficient enrichment of precisely genetically engineered cells is urgently required. Here, we screen for mutations in the receptor for Diphtheria Toxin (DT) which protect human cells from DT. Selection for cells with an edited DT receptor variant enriches for simultaneously introduced, precisely targeted gene modifications at a second independent locus, such as nucleotide substitutions and DNA insertions. Our method enables the rapid generation of a homogenous cell population with bi-allelic integration of a DNA cassette at the selection locus, without clonal isolation. Toxin-based selection works in both cancer-transformed and non-transformed cells, including human induced pluripotent stem cells and human primary T-lymphocytes, as well as it is applicable also in vivo, in mice with humanized liver. This work represents a flexible, precise, and efficient selection strategy to engineer cells using CRISPR-Cas and base editing systems., Genome engineering in cell lines or human stem cells often has poor efficiency, limiting the development of research and therapeutic applications. Here, the authors use a toxin-based selection system for precise bi-allelic engineering in cells and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
12. Potent and selective aldo-keto reductase 1C3 (AKR1C3) inhibitors based on the benzoisoxazole moiety: application of a bioisosteric scaffold hopping approach to flufenamic acid
- Author
-
Davide Bonanni, Marco Lucio Lolli, Weixiao Yuan Wahlgren, Rosmarie Friemann, Renzo Bagnati, Daniele Zonari, Agnese Chiara Pippione, Simonetta Oliaro-Bosso, Donatella Boschi, Parveen Goyal, Salvatore Adinolfi, Claudio Festuccia, Klaus Pors, Elisabetta Marini, Irene Maria Carnovale, and Marcella Sini
- Subjects
17β-HSD5 ,0301 basic medicine ,Aldo-keto reductase 1C3 ,Triazole ,Antineoplastic Agents ,Reductase ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Moiety ,Enzalutamide ,Testosterone ,CRPC ,Enzyme Inhibitors ,Cell Proliferation ,Prostate cancer (PCa) ,X-ray crystallography ,Bioisosterism ,Pharmacology ,Benzoxazoles ,Aldo-keto reductase ,Scaffold hopping ,Dose-Response Relationship, Drug ,Molecular Structure ,Inhibitors ,Chemistry ,Cell growth ,Organic Chemistry ,Aldo-Keto Reductase Family 1 Member C3 ,General Medicine ,Prostate-Specific Antigen ,AKR1C3 ,Flufenamic Acid ,030104 developmental biology ,Flufenamic acid ,Biochemistry ,030220 oncology & carcinogenesis ,Aldo-keto reductase 1C3, AKR1C3, 17β-HSD5, Prostate cancer (PCa), CRPC, Bioisosterism, Scaffold hopping, Inhibitors, X-ray crystallography ,Drug Screening Assays, Antitumor ,Pharmacophore ,medicine.drug - Abstract
The aldo-keto reductase 1C3 (AKR1C3) isoform plays a vital role in the biosynthesis of androgens and is considered an attractive target in prostate cancer (PCa). No AKR1C3-targeted agent has to date been approved for clinical use. Flufenamic acid and indomethacine are non-steroidal anti-inflammatory drugs known to inhibit AKR1C3 in a non-selective manner as COX off-target effects are also observed. Recently, we employed a scaffold hopping approach to design a new class of potent and selective AKR1C3 inhibitors based on a N-substituted hydroxylated triazole pharmacophore. Following a similar strategy, we designed a new series focused around an acidic hydroxybenzoisoxazole moiety, which was rationalised to mimic the benzoic acid role in the flufenamic scaffold. Through iterative rounds of drug design, synthesis and biological evaluation, several compounds were discovered to target AKR1C3 in a selective manner. The most promising compound of series (6) was found to be highly selective (up to 450-fold) for AKR1C3 over the 1C2 isoform with minimal COX1 and COX2 off-target effects. Other inhibitors were obtained modulating the best example of hydroxylated triazoles we previously presented. In cell-based assays, the most promising compounds of both series reduced the cell proliferation, prostate specific antigen (PSA) and testosterone production in AKR1C3-expressing 22RV1 prostate cancer cells and showed synergistic effect when assayed in combination with abiraterone and enzalutamide. Structure determination of AKR1C3 co-crystallized with one representative compound from each of the two series clearly identified both compounds in the androstenedione binding site, hence supporting the biochemical data.
- Published
- 2018
- Full Text
- View/download PDF
13. Rat-Derived Amniotic Epithelial Cells Differentiate into Mature Hepatocytes In Vivo with No Evidence of Cell Fusion
- Author
-
Antonella Contini, Ezio Laconi, Fabio Marongiu, Stephen C. Strom, Maria Paola Serra, Marcella Sini, and Michela Marongiu
- Subjects
Liver cytology ,Biology ,Dipeptidyl peptidase ,Green fluorescent protein ,Original Research Reports ,Pregnancy ,Placenta ,medicine ,Animals ,Humans ,Amnion ,Cell fusion ,Stem Cells ,Cell Differentiation ,Epithelial Cells ,Cell Biology ,Hematology ,Molecular biology ,Rats ,Transplantation ,medicine.anatomical_structure ,Liver ,Amniotic epithelial cells ,Hepatocyte ,Immunology ,Hepatocytes ,Female ,Stem Cell Transplantation ,Developmental Biology - Abstract
Amniotic epithelial cells (AEC) derived from human placenta represent a useful and noncontroversial source for liver-based regenerative medicine. Previous studies suggested that human- and rat-derived AEC differentiate into hepatocyte-like cells upon transplantation. In the retrorsine (RS) model of liver repopulation, clusters of donor-derived cells engrafted in the recipient liver and, importantly, showed characteristics of mature hepatocytes. The aim of the current study was to investigate the possible involvement of cell fusion in the emergence of hepatocyte clusters displaying a donor-specific phenotype. To this end, 4-week-old GFP(+)/DPP-IV(-) rats were treated with RS and then transplanted with undifferentiated AEC isolated from the placenta of DPP-IV(+) pregnant rats at 16-19 days of gestational age. Results indicated that clusters of donor-derived cells were dipeptidyl peptidase type IV (DPP-IV) positive, but did not express the green fluorescent protein (GFP), suggesting that rat amniotic epithelial cells (rAEC) did not fuse within the host parenchyma, as no colocalization of the two tags was observed. Moreover, rAEC-derived clusters expressed markers of mature hepatocytes (eg, albumin, cytochrome P450), but were negative for the expression of biliary/progenitor markers (eg, epithelial cell adhesion molecule [EpCAM]) and did not express the marker of preneoplastic hepatic nodules glutathione S-transferase P (GST-P). These results extend our previous findings on the potential of AEC to differentiate into mature hepatocytes and suggest that this process can occur in the absence of cell fusion with host-derived cells. These studies support the hypothesis that amnion-derived epithelial cells can be an effective cell source for the correction of liver disease.
- Published
- 2015
- Full Text
- View/download PDF
14. Hepatocyte senescence induced by radiation and partial hepatectomy in rat liver
- Author
-
Sarah Koenig, Antonella Contini, Ezio Laconi, Michela Marongiu, Maria Paola Serra, Marcella Sini, Fabio Marongiu, Margret Rave-Fränk, Petra Krause, and Hendrik A. Wolff
- Subjects
Male ,Senescence ,Pathology ,medicine.medical_specialty ,Cell ,Partial hepatectomy ,Biology ,Radiation Dosage ,In vivo ,Parenchyma ,medicine ,Animals ,Hepatectomy ,Radiology, Nuclear Medicine and imaging ,Cells, Cultured ,Cellular Senescence ,Cell Proliferation ,Radiological and Ultrasound Technology ,Kinase ,Rats, Inbred F344 ,Rats ,medicine.anatomical_structure ,Liver ,Rat liver ,Hepatocyte ,Hepatocytes ,Whole-Body Irradiation - Abstract
Exposure to radiation primes the liver for extensive replacement of the resident parenchymal cells by transplanted hepatocytes. The mechanisms underlying this repopulation remain to be clarified. In these studies, we examined the possible occurrence of cell senescence in vivo following radiation-associated preconditioning of the host liver.Fischer 344 rats underwent external-beam, computed-tomography-based partial liver irradiation. A single dose of 25 Gy was delivered to the right liver lobes (40% of liver mass). An additional group of animals received a 1/3 partial hepatectomy (removal of the left anterior lobe) four days after irradiation. Non-irradiated groups served as controls. All rats were sacrificed four weeks after the initial treatment.The irradiated livers displayed several markers of cell senescence, including expression of senescence-associated-β-galactosidase (SA-β-gal), increase in cell size, and up-regulation of cyclin-dependent kinase inhibitors (CDK-I) p16 and p21. Furthermore, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) analysis revealed activation of the senescence-associated secretory phenotype (SASP), including the cytokines interleukin 6 (IL6) and 1α (IL1α). The senescence-related changes were more prominent in rats undergoing partial hepatectomy (PH) following irradiation (IR).We conclude that priming with radiation for liver repopulation results in the induction of cell senescence and the up-regulation of a senescence-associated secretory phenotype. The latter can contribute to the extensive growth of transplanted cells in this system.
- Published
- 2014
- Full Text
- View/download PDF
15. Clearance of senescent hepatocytes in a neoplastic-prone microenvironment delays the emergence of hepatocellular carcinoma
- Author
-
Fabio Marongiu, Maria Paola Serra, Marcella Sini, Ezio Laconi, and Fabrizio Angius
- Subjects
Male ,Senescence ,Aging ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Time Factors ,Context (language use) ,Biology ,medicine.disease_cause ,Tumor Microenvironment ,medicine ,Animals ,Diethylnitrosamine ,cell competition ,cell transplantation ,Cellular Senescence ,Tumor microenvironment ,Interleukin-6 ,Liver Neoplasms ,Cell Biology ,medicine.disease ,Rats, Inbred F344 ,Rats ,Transplantation ,Disease Models, Animal ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Cellular Microenvironment ,liver carcinogenesis ,cell senescence ,Hepatocellular carcinoma ,Hepatocyte ,Hepatocytes ,Carcinogenesis ,Cell aging ,Biomarkers ,liver repopulation ,Research Paper - Abstract
Increasing evidence indicates that carcinogenesis is dependent on the tissue context in which it occurs, implying that the latter can be a target for preventive or therapeutic strategies. We tested the possibility that re-normalizing a senescent, neoplastic-prone tissue microenvironment would exert a modulatory effect on the emergence of neoplastic disease. Rats were exposed to a protocol for the induction of hepatocellular carcinoma (HCC). Using an orthotopic and syngeneic system for cell transplantation, one group of animal was then delivered 8 million normal hepatocytes, via the portal circulation. Hepatocytes transplantation resulted in a prominent decrease in the incidence of both pre-neoplastic and neoplastic lesions. At the end of 1 year 50% of control animals presented with HCC, while no HCC were observed in the transplanted group. Extensive hepatocyte senescence was induced by the carcinogenic protocol in the host liver; however, senescent cells were largely cleared following infusion of normal hepatocytes. Furthermore, levels of Il-6 increased in rats exposed to the carcinogenic protocol, while they returned to near control values in the group receiving hepatocyte transplantation. These results support the concept that strategies aimed at normalizing a neoplastic-prone tissue landscape can modulate progression of neoplastic disease.
- Published
- 2014
- Full Text
- View/download PDF
16. Aging promotes neoplastic disease through effects on the tissue microenvironment
- Author
-
Erika Cadoni, Silvia Doratiotto, Maura Fanti, Marcella Sini, Ezio Laconi, Mônica Campos Serra, Maria Paola Serra, and Fabio Marongiu
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Aging ,selection ,Biology ,pre-neoplastic hepatocytes ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Liver Neoplasms, Experimental ,Liver nodules ,medicine ,Animals ,Dipeptidyl peptidase-4 ,chemistry.chemical_classification ,Messenger RNA ,Neoplastic disease ,Cancer ,Cell Biology ,Visible Lesion ,medicine.disease ,microenvironment ,Rats, Inbred F344 ,Rats ,030104 developmental biology ,Enzyme ,chemistry ,Liver ,liver carcinogenesis ,030220 oncology & carcinogenesis ,Hepatocytes ,Carcinogenesis ,Precancerous Conditions ,Research Paper - Abstract
A better understanding of the complex relationship between aging and cancer will provide important tools for the prevention and treatment of neoplasia. In these studies, the hypothesis was tested that aging may fuel carcinogenesis via alterations imposed in the tissue microenvironment. Preneoplastic hepatocytes isolated from liver nodules were orthotopically injected into either young or old syngeneic rats and their fate was followed over time using the dipeptidyl-peptidase type IV (DPPIV) system to track donor-derived-cells. At 3 months post-Tx, the mean size of donor-derived clusters was 11±3 cells in young vs. 42±8 in old recipients. At 8 months post-Tx, no visible lesion were detected in any of 21 young recipients, while 17/18 animals transplanted at old age displayed hepatic nodules, including 7 large tumors. All tumors expressed the DPPIV marker enzyme, indicating that they originated from transplanted cells. Expression of senescence-associated β-galactosidase was common in liver of 18-month old animals, while it was a rare finding in young controls. Finally, both mRNA and IL6 protein were found to be increased in the liver of aged rats compared to young controls. These results are interpreted to indicate that the microenvironment of the aged liver promotes the growth of pre-neoplastic hepatocytes.
- Published
- 2016
17. Hepatocyte senescence in vivo following preconditioning for liver repopulation
- Author
-
Ezio Laconi, Maria Paola Serra, Marcella Sini, and Fabio Marongiu
- Subjects
Male ,Senescence ,medicine.medical_specialty ,Transplantation Conditioning ,Cell Transplantation ,DNA damage ,Cell ,Biology ,Internal medicine ,medicine ,Animals ,Cellular Senescence ,Pyrrolizidine Alkaloids ,Hepatology ,Interleukin-6 ,Cell Cycle ,Cell cycle ,beta-Galactosidase ,Antineoplastic Agents, Phytogenic ,Rats, Inbred F344 ,Liver regeneration ,Liver Regeneration ,Rats ,Cell biology ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Liver ,Hepatocyte ,Hepatocytes ,Cell aging ,Cell Division ,DNA Damage - Abstract
In the retrorsine (RS)-based model of massive liver repopulation, preexposure to this naturally occurring alkaloid is sufficient to prime normal host parenchymal cells to be slowly replaced by transplanted normal hepatocytes. The basis for this striking effect is yet to be fully elucidated. In the present studies the possible involvement of cell senescence was investigated. Fischer 344 rats were treated according to the RS-based protocol for hepatocyte transplantation, i.e., two doses of RS, 2 weeks apart, and were killed at 4 or 8 weeks after treatment. Control groups were given saline. Expression of senescence-associated beta-galactosidase was greatly induced in hepatocytes exposed to RS. In addition, several other changes that have been related to cell senescence were observed: these included markers of persistent activation of a DNA damage response, an increased expression of mammalian target of rapamycin, and positive regulators of the cell cycle, together with the induction of p21 and p27 cyclin-dependent kinase inhibitors. Furthermore, RS treatment increased levels of interleukin-6 in the liver, consistent with the activation of a senescenceassociated secretory phenotype. Conclusion: These findings indicate that RS induces hepatocyte senescence in vivo. We propose that cell senescence and the associated secretory phenotype can contribute to the selective growth of transplanted hepatocytes in this system. (HEPATOLOGY 2012;56:760-768)
- Published
- 2012
- Full Text
- View/download PDF
18. Hepatic differentiation of amniotic epithelial cells
- Author
-
Kristen J. Skvorak, Ronald N. Hines, Keitaro Mitamura, Shringi Sharma, Fabio Marongiu, Ezio Laconi, Toshio Miki, Marcella Sini, Tiffany L. Sellaro, Roberto Gramignoli, Kenneth Dorko, Aarati Ranade, Ewa Ellis, Stephen C. Strom, Silvia Doratiotto, Stephen F. Badylak, Julio C. Davila, Maria Paola Serra, and Veysel Tahan
- Subjects
Pathology ,medicine.medical_specialty ,Amniotic fluid ,Hepatology ,Cellular differentiation ,Cell Differentiation ,Epithelial Cells ,Amniotic stem cells ,Biology ,Embryonic stem cell ,Article ,Cell biology ,Mice, Inbred C57BL ,Transplantation ,Mice ,medicine.anatomical_structure ,Amniotic epithelial cells ,Hepatocyte ,Hepatocytes ,medicine ,Animals ,Humans ,Syngenic ,Amnion ,Cells, Cultured - Abstract
Hepatocyte transplantation to treat liver disease is largely limited by the availability of useful cells. Human amniotic epithelial cells (hAECs) from term placenta express surface markers and gene characteristics of embryonic stem cells and have the ability to differentiate into all three germ layers, including tissues of endodermal origin (i.e., liver). Thus, hAECs could provide a source of stem cell–derived hepatocytes for transplantation. We investigated the differentiation of hAECs in vitro and after transplantation into the livers of severe combined immunodeficient (SCID)/beige mice. Moreover, we tested the ability of rat amniotic epithelial cells (rAECs) to replicate and differentiate upon transplantation into a syngenic model of liver repopulation. In vitro results indicate that the presence of extracellular matrix proteins together with a mixture of growth factors, cytokines, and hormones are required for differentiation of hAECs into hepatocyte-like cells. Differentiated hAECs expressed hepatocyte markers at levels comparable to those of fetal hepatocytes. They were able to metabolize ammonia, testosterone, and 17α-hydroxyprogesterone caproate, and expressed inducible fetal cytochromes. After transplantation into the liver of retrorsine (RS)-treated SCID/beige mice, naive hAECs differentiated into hepatocyte-like cells that expressed mature liver genes such as cytochromes, plasma proteins, transporters, and other hepatic enzymes at levels equal to adult liver tissue. When transplanted in a syngenic animal pretreated with RS, rAECs were able to engraft and generate a progeny of cells with morphology and protein expression typical of mature hepatocytes. Conclusion: Amniotic epithelial cells possess the ability to differentiate into cells with characteristics of functional hepatocytes both in vitro and in vivo, thus representing a useful and noncontroversial source of cells for transplantation. (HEPATOLOGY 2011;)
- Published
- 2011
- Full Text
- View/download PDF
19. Dual-Specificity Phosphatase 1 Ubiquitination in Extracellular Signal-Regulated Kinase–Mediated Control of Growth in Human Hepatocellular Carcinoma
- Author
-
Maria Maddalena Simile, R Pellegrino, Maria Lauda Tomasi, Lucia Daino, Diego F. Calvisi, Marcella Sini, Francesco Feo, Federico Pinna, Maddalena Frau, Patrizia Virdis, Sara Ladu, Maria Antonietta Seddaiu, Floriana Meloni, Maria Rosaria De Miglio, Rosa Maria Pascale, and Maria Rosaria Muroni
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cyclin-dependent kinase 1 ,Carcinoma, Hepatocellular ,Proliferation index ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Ubiquitin ,Kinase ,Blotting, Western ,Liver Neoplasms ,Dual Specificity Phosphatase 1 ,Molecular biology ,Enzyme Activation ,Oncology ,Ubiquitin ligase complex ,Dual-specificity phosphatase ,biology.protein ,Cancer research ,SKP2 ,Humans ,Immunoprecipitation ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase A ,Cell Division - Abstract
Sustained activation of extracellular signal-regulated kinase (ERK) has been detected previously in numerous tumors in the absence of RAS-activating mutations. However, the molecular mechanisms responsible for ERK-unrestrained activity independent of RAS mutations remain unknown. Here, we evaluated the effects of the functional interactions of ERK proteins with dual-specificity phosphatase 1 (DUSP1), a specific inhibitor of ERK, and S-phase kinase-associated protein 2 (SKP2)/CDC28 protein kinase 1b (CKS1) ubiquitin ligase complex in human hepatocellular carcinoma (HCC). Levels of DUSP1, as assessed by real-time reverse transcription–PCR and Western blot analysis, were significantly higher in tumors with better prognosis (as defined by the length of patients' survival) when compared with both normal and nontumorous surrounding livers, whereas DUSP1 protein expression sharply declined in all HCC with poorer prognosis. In the latter HCC subtype, DUSP1 inactivation was due to either ERK/SKP2/CKS1-dependent ubiquitination or promoter hypermethylation associated with loss of heterozygosity at the DUSP1 locus. Noticeably, expression levels of DUSP1 inversely correlated with those of activated ERK, as well as with proliferation index and microvessel density, and directly with apoptosis and survival rate. Subsequent functional studies revealed that DUSP1 reactivation led to suppression of ERK, CKS1, and SKP2 activity, inhibition of proliferation and induction of apoptosis in human hepatoma cell lines. Taken together, the present data indicate that ERK achieves unrestrained activity during HCC progression by triggering ubiquitin-mediated proteolysis of its specific inhibitor DUSP1. Thus, DUSP1 may represent a valuable prognostic marker and ERK, CKS1, or SKP2 potential therapeutic targets for human HCC. [Cancer Res 2008;68(11):4192–200]
- Published
- 2008
- Full Text
- View/download PDF
20. SKP2 cooperates with N-Ras or AKT to induce liver tumor development in the mouse
- Author
-
Diego F. Calvisi, Xin Chen, Thomas Longerich, Antonio Cigliano, Marcella Sini, Salvatore Delogu, Silvia Ribback, Matthias Evert, Kirsten Utpatel, Frank Dombrowski, and Nina Waldburger
- Subjects
medicine.medical_specialty ,Liver tumor ,Endocrinology ,Internal medicine ,Gastroenterology ,medicine ,Cancer research ,SKP2 ,Biology ,medicine.disease ,Protein kinase B - Published
- 2015
- Full Text
- View/download PDF
21. Co-activation of PIK3CA and Yap promotes development of hepatocellular and cholangiocellular tumors in mouse and human liver
- Author
-
Frank Dombrowski, Matthias Evert, Chunmei Wang, Julien Calderaro, Maria I. Demartis, Diego F. Calvisi, Kirsten Utpatel, Xiaolei Li, Xin Chen, Antonio Cigliano, Marcella Sini, Daniel Azoulay, Katja Evert, Yan Liu, Silvia Ribback, Junyan Tao, and Lijie Jiang
- Subjects
MAPK/ERK pathway ,Pathology ,medicine.medical_specialty ,liver tumor ,Liver tumor ,Carcinoma, Hepatocellular ,Class I Phosphatidylinositol 3-Kinases ,Cell Cycle Proteins ,mTORC1 ,Biology ,PI3K ,Cholangiocarcinoma ,Mice ,Phosphatidylinositol 3-Kinases ,Liver Neoplasms, Experimental ,Cell Line, Tumor ,medicine ,Animals ,Humans ,HCC ,neoplasms ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cell growth ,Liver Neoplasms ,YAP-Signaling Proteins ,Transfection ,medicine.disease ,Phosphoproteins ,Oncology ,Hepatocellular carcinoma ,Cancer research ,Signal transduction ,hippo ,Signal Transduction ,Transcription Factors ,Research Paper - Abstract
Activation of the PI3K and Yes-associated protein (Yap) signaling pathways has been independently reported in human hepatocellular carcinoma (HCC). However, the oncogenic interactions between these two cascades in hepatocarcinogenesis remain undetermined. To assess the consequences of the crosstalk between the PI3K and Yap pathways along liver carcinogenesis, we generated a mouse model characterized by combined overexpression of activated mutant forms of PIK3CA (PIK3CAH1047R) and Yap (YapS127A) in the mouse liver using hydrodynamic transfection (PIK3CA/Yap). In addition, suppression of PI3K and Yap pathways was conducted in human HCC and cholangiocarcinoma (CCA) cell lines. We found that concomitant activation of PI3K and Yap pathways triggered rapid liver tumor development in mice. Histologically, tumors were pure HCC, CCA, or mixed HCC/CCA. At the molecular level, PIK3CA/Yap tumors were characterized by activation of the mTORC1/2, ERK/MAPK, and Notch pathways. Simultaneous activation of PI3K and Yap pathways frequently occurred in human liver tumor specimens and their combined suppression was highly detrimental for the growth of HCC and CCA cell lines. In conclusion, our study demonstrates the oncogenic cooperation between PI3K and Yap pathways along liver carcinogenesis. The PIK3CA/Yap mouse represents an important preclinical liver tumor model for the development of novel therapeutics against this malignancy.
- Published
- 2015
22. 4EBP1/eIF4E and p70S6K/RPS6 axes play critical and distinct roles in hepatocarcinogenesis driven by AKT and N-Ras proto-oncogenes in mice
- Author
-
Jeffrey W. Smith, Xin Chen, Lijie Jiang, Biao Fan, Yan Liu, Frank Dombrowski, Matthias Evert, Maria G. Pilo, Bing Gui, Diego F. Calvisi, Xiaolei Li, Antonio Cigliano, Marcella Sini, and Chunmei Wang
- Subjects
Cell Cycle Proteins ,Apoptosis ,mTORC1 ,Medical Biochemistry and Metabolomics ,Proto-Oncogene Mas ,Mice ,Liver Neoplasms, Experimental ,Anti-apoptotic Ras signalling cascade ,2.1 Biological and endogenous factors ,Pyrophosphatases ,Eukaryotic Initiation Factors ,Aetiology ,Cancer ,Oncogene Proteins ,Ribosomal Protein S6 ,Tumor ,TOR Serine-Threonine Kinases ,Liver Disease ,EIF4E ,Liver Neoplasms ,Ribosomal Protein S6 Kinases, 70-kDa ,Adaptor Proteins ,Ribosomal protein s6 ,Ectonucleoside triphosphate diphosphohydrolase 5 ,Liver Cancer ,Carcinoma, Hepatocellular ,Clinical Sciences ,Immunology ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Cell Line ,Proto-Oncogene Proteins p21(ras) ,Experimental ,Rare Diseases ,Cell Line, Tumor ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Sirolimus ,Hepatology ,Gastroenterology & Hepatology ,Ribosomal Protein S6 Kinases ,Carcinoma ,Signal Transducing ,Hepatocellular ,Phosphoproteins ,70-kDa ,Eukaryotic Initiation Factor-4E ,Multiprotein Complexes ,Cancer research ,Carrier Proteins ,Digestive Diseases ,Proto-Oncogene Proteins c-akt - Abstract
Concomitant expression of activated forms of v-akt murine thymoma viral oncogene homolog (AKT) and Ras in mouse liver (AKT/Ras) leads to rapid tumor development through strong activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway. mTORC1 functions by regulating p70S6K/ribosomal protein S6 (RPS6) and eukaryotic translation initiation factor 4E-binding protein 1/ eukaryotic translation initiation factor 4E (4EBP1/eIF4E) cascades. How these cascades contribute to hepatocarcinogenesis remains unknown. Here, we show that inhibition of the RPS6 pathway by rapamycin effectively suppressed, whereas blockade of the 4EBP1/eIF4E cascade by 4EBP1A4, an unphosphorylatable form of 4EBP1, significantly delayed, AKT/Ras-induced hepatocarcinogenesis. Combined treatment with rapamycin and 4EBP1A4 completely inhibited AKT/Ras hepatocarcinogenesis. This strong antineoplastic effect was successfully recapitulated by ablating regulatory associated protein of mTORC1, the major subunit of mTORC1, in AKT/Ras-overexpressing livers. Furthermore, we demonstrate that overexpression of eIF4E, the proto-oncogene whose activity is specifically inhibited by 4EBP1, resulted in hepatocellular carcinoma (HCC) development in cooperation with activated Ras. Mechanistically, we identified the ectonucleoside triphosphate diphosphohydrolase 5/ adenylate kinase 1/cytidine monophosphate kinase 1 axis and the mitochondrial biogenesis pathway as targets of the 4EBP1/eIF4E cascade in AKT/Ras and Ras/eIF4E livers as well as in human HCC cell lines and tissues. Conclusions: Complete inhibition of mTORC1 is required to suppress liver cancer development induced by AKT and Ras proto-oncogenes in mice. The mTORC1 effectors, RPS6 and eIF4E, play distinct roles and are both necessary for AKT/Ras hepatocarcinogenesis. These new findings might open the way for innovative therapies against human HCC. (Hepatology 2015;61:200–213)
- Published
- 2015
23. Advances and Possible Applications of Human Amnion for the Management of Liver Disease
- Author
-
Marc C. Hansel, Stephen C. Strom, Kristen J. Skvorak, Roberto Gramignoli, Fabio Marongiu, Maria Paola Serra, Ezio Laconi, and Marcella Sini
- Subjects
Liver disease ,Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,Amnion ,business.industry ,medicine ,medicine.disease ,business - Published
- 2013
- Full Text
- View/download PDF
24. Abstract 1270: Polysialyltransferase ST8SiaII as a target for neuroblastoma dissemination
- Author
-
Rida F. Saeed, Sara M. Elkashef, Xiaoxiao Guo, Robert A. Falconer, Steven D. Shnyder, Mark Sutherland, Goreti Ribeiro Morais, Laurence H. Patterson, Paul M. Loadman, Virginie Viprey, and Marcella Sini
- Subjects
Cancer Research ,biology ,Polysialic acid ,Chemistry ,Sialyltransferase ,Cell migration ,medicine.disease ,01 natural sciences ,Isogenic human disease models ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Oncology ,Neuroblastoma ,Cancer research ,medicine ,biology.protein ,Neural cell adhesion molecule ,Neuronal Cell Adhesion Molecule ,Protein kinase B - Abstract
Polysialic acid (polySia) is expressed on the surface of NCAM (neuronal cell adhesion molecule) on neuroendocrine tumours, notably neuroblastoma and small cell lung cancer, where it modulates cell-cell and cell-matrix adhesion, migration, invasion and metastasis. PolySia expression is strongly associated with poor prognosis and aggressive disease in neuroblastoma patients in the clinic[1]. SiRNA knockdown of polysialyltransferase (polyST) ST8SiaII, the enzyme primarily responsible for polySia synthesis in tumours, abrogates tumour cell migration and invasion. Besides brain regions with persistent neuronal plasticity, polySia is essentially absent from the body post-embryogenesis. PolyST is thus a selective and largely unexplored therapeutic target for neuroblastoma dissemination [1]. We have established a highly sensitive HPLC-based polyST inhibition assay, amenable to high-throughput screening. We report our efforts to further optimise this cell-free assay, and include details of our novel methodology to quantify cell-surface polySia expression. Having demonstrated in vitro that inhibition of polyST by a small molecule leads to a reduction in tumour cell migration [2], we designed and synthesised ST8SiaII inhibitors. Using isogenic cell lines (C6-STX: polySia+/polyST+ and C6-WT: polySia-/polyST-) and naturally polySia expressing human neuroblastoma cells (SH-SY5Y, IMR-32) these compounds were evaluated for their ability to reduce polySia expression, to modulate tumour cell migration and invasion in vitro. We have identified novel agents which significantly reduce polySia expression, tumour cell migration and invasion. These effects were only found in cell lines expressing ST8SiaII and polySia. Specificity of agents for polySTs over other members of the sialyltransferase (ST) family (i.e. α-2,3-ST and α-2,6-ST) was subsequently investigated using lectin differential labelling probes. Agents did not inhibit sialyltransferase activity. We have investigated effects of agents on key intracellular signalling pathways. We demonstrated the effects of polyST inhibition on the dynamics of FAK and on ERK1/2, AKT, CREB and VEGFR-3 signalling. Furthemore, we have explored the behaviour of polySia-expressing cells under hypoxic conditions. Our data suggest that polySia is associated with a resistant phenotype, with C6-STX polySia-expressing cells demonstrating a survival advantage and additionally maintaining their migratory capacity under hypoxia (compared to WT cells, where migration is dramatically reduced). In summary, we have demonstrated that polyST inhibition dramatically decreases cell-surface polysialylation, migration and invasion in vitro, under both normoxic and hypoxic conditions. This work paves the way for development of a novel therapeutic for the treatment of neuroblastoma. [1] Falconer, R.A. et al., Curr. Cancer Drug Targets, 2012, 12, 925-939; [2] Al-Saraireh YMJ et al., PLoS ONE, 2013, 8:e73366. Citation Format: Sara M. Elkashef, Rida F. Saeed, Goreti Ribeiro Morais, Xiaoxiao Guo, Marcella Sini, Virginie F. Viprey, Mark Sutherland, Paul M. Loadman, Laurence H. Patterson, Steven D. Shnyder, Robert A. Falconer. Polysialyltransferase ST8SiaII as a target for neuroblastoma dissemination. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1270.
- Published
- 2016
- Full Text
- View/download PDF
25. Cancer as a disease of tissue pattern formation
- Author
-
Silvia Doratiotto, Fabio Marongiu, Maria Paola Serra, Marcella Sini, and Ezio Laconi
- Subjects
Tumor microenvironment ,Pathology ,medicine.medical_specialty ,Histology ,Clinical Biochemistry ,Morphogenesis ,Cancer ,Cell Biology ,Disease ,Biology ,medicine.disease ,Pathogenesis ,Neoplasms ,Cell polarity ,medicine ,Bystander effect ,Tumor Microenvironment ,Humans ,Hypoxia ,Neuroscience ,Process (anatomy) ,Signal Transduction - Abstract
The diagnosis of neoplastic disease still lays its foundations on the detection of altered tissue morphology. Most importantly, cancer begins, at least in many cases as a disease with altered tissue pattern formation. It is therefore rather surprising that the issue regarding the possible mechanistic role of such property in the pathogenesis of cancer has received relatively little attention so far. To be more specific, we need to ask the following question: is altered tissue pattern formation a mere bystander, with its pervasive presence along the entire carcinogenic sequence, or does it play a role in fuelling this process? Pathways related to morphogenesis and to the establishment of cell polarity will be considered for their possible mechanistic involvement in early phases of neoplastic disease. Evidences and hypotheses relating altered tissue pattern formation to the emergence of the tumor microenvironment and to neoplastic progression will be discussed.
- Published
- 2012
26. Murine double minute 2 regulates Hu antigen R stability in human liver and colon cancer through NEDDylation
- Author
-
Ana Belén de la Hoz, Marta Varela-Rey, Virginia Gutiérrez-de Juan, Nieves Embade, Dimitris P. Xirodimas, Francisco Javier Bustamante, Nuria Martinez-Lopez, Naiara Beraza, David Fernández-Ramos, Begoña Rodríguez-Iruretagoyena, Marcella Sini, Arkaitz Carracedo, María L. Martínez-Chantar, José M. Mato, Shelly C. Lu, Manuel S. Rodriguez, and Ashwin Woodhoo
- Subjects
Male ,Cytoplasm ,Carcinoma, Hepatocellular ,NEDD8 Protein ,medicine.disease_cause ,NEDD8 ,Article ,Protein neddylation ,Mice ,Proto-Oncogene Proteins c-mdm2 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Ubiquitins ,Cell Nucleus ,Hepatology ,biology ,Liver Neoplasms ,MRNA stabilization ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Cell Transformation, Neoplastic ,ELAV Proteins ,Case-Control Studies ,Colonic Neoplasms ,biology.protein ,Hepatocytes ,Mdm2 ,Neddylation ,Carcinogenesis ,Cullin ,Signal Transduction - Abstract
HuR, a member of the Elav/Hu family, is a gene expression regulator with a central role in mRNA stabilization (1), HuR has been shown to increase cellular division by enhancing the stability of mRNAs encoding important proteins for cell cycle control and proliferation, as well as other factors that influence tumor cell growth (2, 3). HuR expression is elevated in colon, breast, prostate, pancreatic, oral, skin, brain, gastric, lung and ovarian cancers, and correlates with tumor malignancy, supporting a role for HuR in tumorigenesis (4–6). In the liver, HuR plays a crucial role in hepatocyte proliferation, differentiation, and HCC transformation (7). At the transcriptional level, NF-κB can activate HuR transcription downstream of the PI3K-AKT signaling pathway (8). At the post-transcriptional level, the ubiquitin (Ub)-proteasome pathway has been implicated in HuR function, whereas the machinery involved in enhancing HuR protein stability remains obscure (9). The ubiquitin-like molecule NEDD8 is a key regulator of cell growth, viability and development. NEDD8 is ubiquitously expressed and highly conserved among most eukaryotes. At the molecular level, only members of the cullin family of proteins, are well-characterized substrates for NEDDylation (10). However, through direct biological approaches or proteomic techniques, it is evident that NEDD8 proteome is widely diverse. Recent studies have shown that ribonucleoproteins are targets for NEDD8 conjugation which protect them from de-stabilization (11, 12). Conversely, cysteine protease (NEDP1) removes NEDD8 molecules from conjugated substrates (13). Mdm2 acts as an E3 NEDD8 ligase, promoting p53 NEDDylation and stabilization (14), and, of importance, Mdm2 is overexpressed in many human tumors, at least in part due to gene amplification (15). In this study, we show that HuR is overexpressed in proliferative HCC and colon cancer cells and biopsies through Mdm2-mediated NEDDylation. Our data suggests that HuR NEDDylation at K313 and K326 residues stabilize HuR. Mechanistically, Mdm2-mediated NEDDylation of HuR controls the nuclear localization of HuR and protects it from degradation. To our knowledge, this is the first report to implicate NEDDylation in the regulation of HuR stability and localization. Our findings show that NEDDylation is a novel mechanism for HuR regulation that might represent a useful tool for new therapeutic strategies for HCC and colon cancer.
- Published
- 2011
27. The degradation of cell cycle regulators by SKP2/CKS1 ubiquitin ligase is genetically controlled in rodent liver cancer and contributes to determine the susceptibility to the disease
- Author
-
Federico Pinna, Maria Maddalena Simile, Maria Rosaria Muroni, Maria Lauda Tomasi, Diego F. Calvisi, Marcella Sini, Rosa Maria Pascale, Francesco Feo, Maria Antonietta Seddaiu, Maddalena Frau, Sara Ladu, and Ilaria Demartis
- Subjects
Cancer Research ,Proliferation index ,Cell Cycle Proteins ,medicine.disease_cause ,Ubiquitin ,medicine ,SKP2 ,Animals ,Humans ,Immunoprecipitation ,Genetic Predisposition to Disease ,S-Phase Kinase-Associated Proteins ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Liver Neoplasms ,Ubiquitination ,Cell cycle ,HCCS ,Immunohistochemistry ,Phenotype ,digestive system diseases ,Rats ,Ubiquitin ligase ,Oncology ,Biochemistry ,biology.protein ,Cancer research ,Carcinogenesis ,Precancerous Conditions ,Protein Processing, Post-Translational - Abstract
Previous work showed a genetic control of cell cycle deregulation during hepatocarcinogenesis. We now evaluated in preneoplastic lesions, dysplastic nodules and hepatocellular carcinoma (HCC), chemically induced in genetically susceptible F344 and resistant Brown Norway (BN) rats, the role of cell cycle regulating proteins in the determination of a phenotype susceptible to HCC development. p21(WAF1), p27(KIP1), p57(KIP2) and p130 mRNA levels increased in fast growing lesions of F344 rats. Lower/no increases occurred in slowly growing lesions of BN rats. A similar behavior of RassF1A mRNA was previously found in the 2 rat strains. However, p21(WAF1), p27(KIP1), p57(KIP), p130 and RassF1A proteins exhibited no change/low increase in the lesions of F344 rats and consistent rise in dysplastic nodules and HCC of BN rats. Increase in Cks1-Skp2 ligase and ubiquitination of cell cycle regulators occurred in F344 but not in BN rat lesions, indicating that posttranslational modifications of cell cycle regulators are under genetic control and contribute to determine a phenotype susceptible to HCC. Moreover, proliferation index of 60 human HCCs was inversely correlated with protein levels but not with mRNA levels of P21(WAF1), P27(KIP1), P57(KIP2) and P130, indicating a control of human HCC proliferation by posttranslational modifications of cell cycle regulators.
- Published
- 2009
- Full Text
- View/download PDF
28. Ras-driven proliferation and apoptosis signaling during rat liver carcinogenesis is under genetic control
- Author
-
Maria Rosaria Muroni, R Pellegrino, Maria Rosaria De Miglio, Vanna Sanna, Maddalena Frau, Diego F. Calvisi, Federico Pinna, Rosa Maria Pascale, Maria Maddalena Simile, Lucia Daino, Maria Lauda Tomasi, Maria Antonietta Seddaiu, Marcella Sini, and Francesco Feo
- Subjects
MAPK/ERK pathway ,Cancer Research ,Programmed cell death ,Tumor suppressor gene ,Apoptosis ,Biology ,medicine.disease_cause ,MAP Kinase Kinase Kinase 5 ,Liver Neoplasms, Experimental ,Rats, Inbred BN ,medicine ,Animals ,Genetic Predisposition to Disease ,Extracellular Signal-Regulated MAP Kinases ,Cell Proliferation ,Cell growth ,Dual Specificity Phosphatase 1 ,digestive system diseases ,Rats, Inbred F344 ,Rats ,Adaptor Proteins, Vesicular Transport ,Oncology ,Mitogen-activated protein kinase ,Cancer research ,biology.protein ,ras Proteins ,Signal transduction ,Carcinogenesis ,Precancerous Conditions ,Signal Transduction - Abstract
Fast growth and deregulation of G1 and S phases characterize preneoplastic and neoplastic liver lesions of genetically susceptible F344 rats, whereas a G1-S block in lesions of resistant BN rats explains their low progression capacity. However, signal transduction pathways responsible for the different propensity of lesions from the 2 rat strains to evolve to malignancy remain unknown. Here, we comparatively investigated the role of Ras/Erk pathway inhibitors, involved in growth restraint and cell death, in the acquisition of a phenotype resistant or susceptible to hepatocarcinogenesis. Moderate activation of Ras, Raf-1 and Mek proteins was paralleled in both rat models by strong induction of Dab2 and Rkip inhibitors. Levels of Dusp1, a specific ERK inhibitor, increased only in BN rat lesions, leading to modest ERK activation, whereas a progressive Dusp1 decline occurred in corresponding lesions from F344 rats and was accompanied by elevated ERK activation. Furthermore, a gradual increase of Rassf1A/Nore1A/Mst1-driven apoptosis was detected in both rat strains, with highest levels in BN hepatocellular carcinoma (HCC), whereas loss of Dab2IP, a protein implicated in ASK1-dependent cell death, occurred only in F344 rat HCC, resulting in significantly higher apoptosis in BN than F344 HCC. Taken together, our results indicate a control of the Ras/Erk pathway and the pro-apoptotic Rassf1A/Nore1A and Dab2IP/Ask1 pathways by HCC susceptibility genes. Dusp1 possesses a prominent role in the acquisition of the phenotype resistant to HCC by BN rats, whereas late activation of RassF1A/Nore1A and Dab2IP/Ask1 axes is implicated in the highest apoptosis characteristic of BN HCC.
- Published
- 2008
29. 432 CELL TURNOVER IN THE REPOPULATED RAT LIVER: EVIDENCE AGAINST HEPATOCYTE STREAMING
- Author
-
Marcella Sini, Mp Serra, Ezio Laconi, and Fabio Marongiu
- Subjects
medicine.anatomical_structure ,Hepatology ,Chemistry ,Hepatocyte ,Rat liver ,Cell turnover ,medicine ,Cell biology - Published
- 2012
- Full Text
- View/download PDF
30. 423 CELL SENESCENCE IN LIVER REPOPULATION AND CARCINOGENESIS
- Author
-
Mp Serra, Fabio Marongiu, Marcella Sini, and Ezio Laconi
- Subjects
Senescence ,medicine.anatomical_structure ,Hepatology ,Cell ,medicine ,Cancer research ,Biology ,Carcinogenesis ,medicine.disease_cause ,Liver repopulation - Published
- 2012
- Full Text
- View/download PDF
31. P9 THE LOSS OF PROLIFERATIVE COMPETITIVENESS OF AGED HEPATOCYTES
- Author
-
Francesco Marongiu, Mp Serra, Marcella Sini, Ezio Laconi, Michela Marongiu, and Antonella Contini
- Subjects
Hepatology - Published
- 2014
- Full Text
- View/download PDF
32. P11 IN VIVO DIFFERENTIATION OF AMNIOTIC EPITHELIAL CELLS INTO MATURE HEPATOCYTES: NO EVIDENCE OF CELL FUSION
- Author
-
Francesco Marongiu, Marcella Sini, Mp Serra, Michela Marongiu, Antonella Contini, and Ezio Laconi
- Subjects
Cell fusion ,Hepatology ,In vivo ,Amniotic epithelial cells ,Amniotic stem cells ,Anatomy ,Biology ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
33. 307 THE MICROENVIRONMENT OF THE AGED RAT LIVER SUPPORTS THE GROWTH OF TRANSPLANTED HEPATOCYTES ISOLATED FROM HEPATIC NODULES
- Author
-
Ezio Laconi, Fabio Marongiu, Marcella Sini, and Mp Serra
- Subjects
Pathology ,medicine.medical_specialty ,Hepatology ,medicine ,Hepatic nodules ,Biology ,Aged rat - Published
- 2012
- Full Text
- View/download PDF
34. In vitro and in vivo differentiation of amniotic epithelial stem cells into hepatocyte-like cells
- Author
-
Shringi Sharma, Ezio Laconi, Mônica Campos Serra, Marcella Sini, Roberto Gramignoli, Silvia Doratiotto, Tiffany L. Sellaro, Fabio Marongiu, Kenneth Dorko, and Stephen C. Strom
- Subjects
Endothelial stem cell ,Reproductive Medicine ,Amniotic epithelial cells ,Neurosphere ,Obstetrics and Gynecology ,Amniotic stem cells ,Stem cell ,Biology ,Cord lining ,Developmental Biology ,Cell biology ,Stem cell transplantation for articular cartilage repair ,Adult stem cell - Published
- 2011
- Full Text
- View/download PDF
35. SKP2 and CKS1 Promote Degradation of Cell Cycle Regulators and Are Associated With Hepatocellular Carcinoma Prognosis
- Author
-
Maria Maddalena Simile, Maria Antonietta Seddaiu, Francesco Feo, Maria Rosaria Muroni, Maddalena Frau, Dae-Sik Lim, Sara Ladu, Federico Pinna, Marcella Sini, Maria Lauda Tomasi, Piero Bonelli, Diego F. Calvisi, and Rosa Maria Pascale
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Small interfering RNA ,Carcinoma, Hepatocellular ,Cell Culture Techniques ,Cell Cycle Proteins ,Biology ,Ubiquitin ,Cyclin-dependent kinase ,Cell Line, Tumor ,CDC2-CDC28 Kinases ,SKP2 ,Humans ,Cell Cycle Protein ,S-Phase Kinase-Associated Proteins ,Hepatology ,Forkhead Box Protein O1 ,Tumor Suppressor Proteins ,Liver Neoplasms ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,Gastroenterology ,Forkhead Transcription Factors ,Cell cycle ,Cyclin-Dependent Kinases ,Ubiquitin ligase ,Cell biology ,biology.protein ,Cancer research ,Anaphase-promoting complex ,Carrier Proteins ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
Background & Aims The cell cycle regulators P21 WAF1 , P27 KIP1 , P57 KIP2 , P130, RASSF1A, and FOXO1 are down-regulated during hepatocellular carcinoma (HCC) pathogenesis. We investigated the role of the ubiquitin ligase subunits CKS1 and SKP2, which regulate proteasome degradation of cell cycle regulators, in HCC progression. Methods Human HCC tissues from patients with better (HCCB, >3 years survival) and poorer prognosis (HCCP, Results The promoters of P21 WAF1 , P27 KIP1 , and P57 KIP2 were more frequently hypermethylated in HCCP than HCCB. Messenger RNA levels of these genes were up-regulated in samples in which these genes were not methylated; protein levels increased only in HCCB because of CKS1- and SKP2-dependent ubiquitination of these proteins in HCCP. The level of SKP2 expression correlated with rate of HCC cell proliferation and level of microvascularization of samples and was inversely correlated with apoptosis and survival. In HCCB, SKP2 activity was balanced by degradation by the ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C)-CDH1 and up-regulation of SKP2 suppressor histidine triad nucleotide binding protein 1 (HINT1). In HCCP, however, SKP2 was not degraded because of down-regulation of the phosphatase CDC14B, CDK2-dependent serine phosphorylation (which inhibits interaction between CDH1 and SKP2), and HINT1 inactivation. In HCC cells, small interfering RNA knockdown of SKP2 reduced proliferation and ubiquitination of the cell cycle regulators, whereas SKP2 increased proliferation and reduced expression of cell cycle regulators. Conclusions Ubiquitination and proteasome degradation of P21WAF1, P27KIP1, P57KIP2, P130, RASSF1A, and FOXO1 and mechanisms that prevent degradation of SKP2 by APC/C-CDH1 contribute to HCC progression. CKS1-SKP2 ligase might be developed as a therapeutic target or diagnostic marker.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.