102 results on '"Pellegata NS"'
Search Results
2. Study on aneuplody nd P53 mutations in astrcytomas
- Author
-
Campomenosi, P, Ottaggio, L, Moro, F, Urbini, S, Bogliolo, M, Zunino, A, Camoirano, A, Inga, A, Gentile, Sl, Pellegata, Ns, Bonassi, S, Bruzzone, E, Iannone, R, Pisani, Roberto, Menichini, P, Ranzani, Gn, Bonatti, S, Abbondandolo, A, and Fronza, G.
- Published
- 1996
3. K-ras and p53 Gene Mutations in Pancreatic Cancer: ductal and non ductal tumors progress through different genetic lesions
- Author
-
Pellegata, Ns, Sessa, Fausto, Renault, B, Bonato, M, Leone, Be, Solcia, E, and Ranzani, Gn
- Published
- 1994
4. Intraductal papillary-mucinous tumours represent a distinct group of pancreatic neoplasm: an investigation of tumor cell differentiation and K-ras, p53 and c.erbB-2 abnormalities in 26 patients
- Author
-
Sessa, Fausto, Solcia, E, Capella, C, Bonato, M, Scarpa, A, Zamboni, G, Pellegata, Ns, Ranzani, Gn, Rickaert, F, and Kloppel, G.
- Published
- 1994
5. Systematic mapping of mitochondrial calcium uniporter channel (MCUC)-mediated calcium signaling networks.
- Author
-
Delgado de la Herran H, Vecellio Reane D, Cheng Y, Katona M, Hosp F, Greotti E, Wettmarshausen J, Patron M, Mohr H, Prudente de Mello N, Chudenkova M, Gorza M, Walia S, Feng MS, Leimpek A, Mielenz D, Pellegata NS, Langer T, Hajnóczky G, Mann M, Murgia M, and Perocchi F
- Subjects
- Humans, Calcium metabolism, Calcium-Binding Proteins metabolism, Calcium-Binding Proteins genetics, HEK293 Cells, Animals, HeLa Cells, Cation Transport Proteins, Mitochondrial Membrane Transport Proteins, Calcium Channels metabolism, Calcium Channels genetics, Calcium Signaling, Mitochondria metabolism
- Abstract
The mitochondrial calcium uniporter channel (MCUC) mediates mitochondrial calcium entry, regulating energy metabolism and cell death. Although several MCUC components have been identified, the molecular basis of mitochondrial calcium signaling networks and their remodeling upon changes in uniporter activity have not been assessed. Here, we map the MCUC interactome under resting conditions and upon chronic loss or gain of mitochondrial calcium uptake. We identify 89 high-confidence interactors that link MCUC to several mitochondrial complexes and pathways, half of which are associated with human disease. As a proof-of-concept, we validate the mitochondrial intermembrane space protein EFHD1 as a binding partner of the MCUC subunits MCU, EMRE, and MCUB. We further show a MICU1-dependent inhibitory effect of EFHD1 on calcium uptake. Next, we systematically survey compensatory mechanisms and functional consequences of mitochondrial calcium dyshomeostasis by analyzing the MCU interactome upon EMRE, MCUB, MICU1, or MICU2 knockdown. While silencing EMRE reduces MCU interconnectivity, MCUB loss-of-function leads to a wider interaction network. Our study provides a comprehensive and high-confidence resource to gain insights into players and mechanisms regulating mitochondrial calcium signaling and their relevance in human diseases., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Opposing Effects of Cannabidiol in Patient-derived Neuroendocrine Tumor, Pheochromocytoma/Paraganglioma Primary Cultures.
- Author
-
Wang K, Schober L, Fischer A, Bechmann N, Maurer J, Peischer L, Reul A, Hantel C, Reincke M, Beuschlein F, Robledo M, Mohr H, Pellegata NS, Schilbach K, Knösel T, Ilmer M, Angele M, Kroiss M, Maccio U, Broglie-Däppen M, Vetter D, Lehmann K, Pacak K, Grossman AB, Auernhammer CJ, Zitzmann K, and Nölting S
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Tumor Cells, Cultured, Aged, Young Adult, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Primary Cell Culture, Cannabidiol pharmacology, Paraganglioma drug therapy, Paraganglioma pathology, Pheochromocytoma drug therapy, Pheochromocytoma pathology, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Adrenal Gland Neoplasms drug therapy, Adrenal Gland Neoplasms pathology
- Abstract
Context: Treatment options for advanced neuroendocrine tumors (NETs), pheochromocytomas and paragangliomas (PPGLs) are still limited. In recent years, antitumor effects of cannabinoids have been reported; however, there are only very limited data available in NETs or PPGLs., Objective: Investigation of the effects of cannabidiol (CBD) on patient-derived human NET/PPGL primary cultures and on NET/PPGL cell lines., Methods: We established primary cultures derived from 46 different patients with PPGLs (n = 35) or NETs (n = 11) who underwent tumor resection at 2 centers. Treatment of patient primary cultures with clinically relevant doses (5 µM) and slightly higher doses (10 µM) of CBD was performed., Results: We found opposing effects of 5 µM CBD: significant antitumor effects in 5/35 (14%) and significant tumor-promoting effects in 6/35 (17%) of PPGL primary cultures. In terms of antitumor effects, cluster 2-related PPGLs showed significantly stronger responsivity to CBD compared to cluster 1-related PPGLs (P = .042). Of the cluster 2-related tumors, NF1 PPGLs showed the strongest responsivity (4/5 PPGL primary cultures with a significant decrease in cell viability were NF1-mutated). We also found opposing effects of 10 µM CBD in PPGLs and NETs: significant antitumor effects in 9/33 of PPGL (27%) and 3/11 of NET (27%) primary cultures and significant tumor-promoting effects in 6/33 of PPGL (18%) and 2/11 of NET (18%) primary cultures., Conclusion: We suggest a potential novel treatment option for some NETs/PPGLs but also provide evidence for caution when applying cannabinoids as supportive therapy for pain or appetite management to cancer patients and possibly as health supplements., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
7. Animal and Cell Culture Models of PPGLs - Achievements and Limitations.
- Author
-
Karna B, Pellegata NS, and Mohr H
- Subjects
- Animals, Humans, Cell Culture Techniques, Paraganglioma metabolism, Pheochromocytoma metabolism, Adrenal Gland Neoplasms metabolism, Neuroendocrine Tumors
- Abstract
Research on rare tumors heavily relies on suitable models for basic and translational research. Paragangliomas (PPGL) are rare neuroendocrine tumors (NET), developing from adrenal (pheochromocytoma, PCC) or extra-adrenal (PGL) chromaffin cells, with an annual incidence of 2-8 cases per million. While most PPGL cases exhibit slow growth and are primarily treated with surgery, limited systemic treatment options are available for unresectable or metastatic tumors. Scarcity of appropriate models has hindered PPGL research, preventing the translation of omics knowledge into drug and therapy development. Human PPGL cell lines are not available, and few animal models accurately replicate the disease's genetic and phenotypic characteristics. This review provides an overview of laboratory models for PPGLs, spanning cellular, tissue, organ, and organism levels. We discuss their features, advantages, and potential contributions to diagnostics and therapeutics. Interestingly, it appears that in the PPGL field, disease models already successfully implemented in other cancers have not been fully explored., Competing Interests: The authors declare that they have no conflict of interest., (Thieme. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Tribbles Genes in Gastric Cancer: A Tumor-Suppressive Role for TRIB2 .
- Author
-
Foscarini A, Tricarico R, Gentile F, Satam S, Mohr H, Kiss-Toth E, Ranzani GN, and Pellegata NS
- Subjects
- Humans, Apoptosis, Cell Line, Tumor, G2 Phase Cell Cycle Checkpoints, Chromosomal Instability, Calcium-Calmodulin-Dependent Protein Kinases, Protein Serine-Threonine Kinases genetics, Intracellular Signaling Peptides and Proteins genetics, Stomach Neoplasms genetics
- Abstract
Tribbles pseudokinases (TRIB1-3) are important signaling modulators involved in several cancers. However, their function in gastric cancer (GC) remains undefined. GC is still a deadly disease since the lack of sensitive and specific biomarkers for early diagnosis and therapy response prediction negatively affects patients' outcome. The identification of novel molecular players may lead to more effective diagnostic and therapeutic avenues. Therefore, we investigated the role of TRIB genes in gastric tumorigenesis. Data mining of the TCGA dataset revealed that chromosomal instability (CIN) tumors have lower TRIB2 and higher TRIB3 expression versus microsatellite instability (MSI)-high tumors, while TRIB1 levels are similar in both tumor types. Moreover, in CIN tumors, low TRIB2 expression is significantly associated with aggressive stage IV disease. As no studies on TRIB2 in GC are available, we focused on this gene for further in vitro analyses. We checked the effect of TRIB2 overexpression (OE) on MKN45 and NCI-N87 CIN GC cell lines. In MKN45 cells, TRIB2 OE reduced proliferation and colony formation ability and induced G2/M arrest, while it decreased the proliferation and cell motility of NCI-N87 cells. These effects were not mediated by the MAPK pathway. Our results suggest a tumor-suppressive function of TRIB2 in GC with a CIN phenotype.
- Published
- 2023
- Full Text
- View/download PDF
9. Multiple endocrine neoplasia type 4 (MEN4): a thorough update on the latest and least known men syndrome.
- Author
-
Ruggeri RM, Benevento E, De Cicco F, Grossrubatscher EM, Hasballa I, Tarsitano MG, Centello R, Isidori AM, Colao A, Pellegata NS, and Faggiano A
- Subjects
- Humans, Syndrome, Multiple Endocrine Neoplasia diagnosis, Multiple Endocrine Neoplasia genetics, Multiple Endocrine Neoplasia pathology, Multiple Endocrine Neoplasia Type 1 genetics, Multiple Endocrine Neoplasia Type 1 diagnosis, Neuroendocrine Tumors genetics, Adenoma genetics
- Abstract
Purpose: Multiple endocrine neoplasia type 4 (MEN4) is a rare multiglandular endocrine neoplasia syndrome, associated with a wide tumor spectrum but hallmarked by primary hyperparathyroidism, which represents the most common clinical feature, followed by pituitary (functional and non-functional) adenomas, and neuroendocrine tumors. MEN4 clinically overlaps MEN type 1 (MEN1) but differs from it for milder clinical features and an older patient's age at onset. The underlying mutated gene, CDKN1B, encodes the cell cycle regulator p27, implicated in cellular proliferation, motility and apoptosis. Given the paucity of MEN4 cases described in the literature, possible genotype-phenotype correlations have not been thoroughly assessed, and specific clinical recommendations are lacking. The present review provides an extensive overview of molecular genetics and clinical features of MEN4, with the aim of contributing to delineate peculiar strategies for clinical management, screening and follow-up of the last and least known MEN syndrome., Methods: A literature search was performed through online databases like MEDLINE and Scopus., Conclusions: MEN4 is much less common that MEN1, tend to present later in life with a more indolent course, although involving the same primary organs as MEN1. As a consequence, MEN4 patients might need specific diagnostic and therapeutic approaches and a different strategy for screening and follow-up. Further studies are needed to assess the real oncological risk of MEN4 carriers, and to establish a standardized screening protocol. Furthermore, a deeper understanding of molecular genetics of MEN4 is needed in order to explore p27 as a novel therapeutic target., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
10. Responses to systemic therapy in metastatic pheochromocytoma/paraganglioma: a retrospective multicenter cohort study.
- Author
-
Fischer A, Kloos S, Remde H, Dischinger U, Pamporaki C, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Bechmann N, Hantel C, Mohr H, Pellegata NS, Bornstein SR, Kroiss M, Auernhammer CJ, Reincke M, Pacak K, Grossman AB, Beuschlein F, and Nölting S
- Subjects
- Humans, Iodine Radioisotopes therapeutic use, Retrospective Studies, Cohort Studies, Temozolomide therapeutic use, Sunitinib therapeutic use, Succinate Dehydrogenase genetics, Somatostatin therapeutic use, Pheochromocytoma pathology, Paraganglioma genetics, Adrenal Gland Neoplasms pathology, Brain Neoplasms, Neoplasms, Second Primary, Cardiovascular Diseases
- Abstract
Objective: The therapeutic options for metastatic pheochromocytomas/paragangliomas (mPPGLs) include chemotherapy with cyclophosphamide/vincristine/dacarbazine (CVD), temozolomide monotherapy, radionuclide therapies, and tyrosine kinase inhibitors such as sunitinib. The objective of this multicenter retrospective study was to evaluate and compare the responses of mPPGLs including those with pathogenic variants in succinate dehydrogenase subunit B (SDHB), to different systemic treatments., Design: This is a retrospective analysis of treatment responses of mPPGL patients (n = 74) to systemic therapies., Methods: Patients with mPPGLs treated at 6 specialized national centers were selected based on participation in the ENSAT registry. Survival until detected progression (SDP) and disease-control rates (DCRs) at 3 months were evaluated based on imaging reports., Results: For the group of patients with progressive disease at baseline (83.8% of 74 patients), the DCR with first-line CVD chemotherapy was 75.0% (n = 4, SDP 11 months; SDHB [n = 1]: DCR 100%, SDP 30 months), with somatostatin peptide receptor-based radionuclide therapy (PPRT) 85.7% (n = 21, SDP 17 months; SDHB [n = 10]: DCR 100%, SDP 14 months), with 131I-meta-iodobenzylguanidine (131I-MIBG) 82.6% (n = 23, SDP 43 months; SDHB [n = 4]: DCR 100%, SDP 24 months), with sunitinib 100% (n = 7, SDP 18 months; SDHB [n = 3]: DCR 100%, SDP 18 months), and with somatostatin analogs 100% (n = 4, SDP not reached). The DCR with temozolomide as second-line therapy was 60.0% (n = 5, SDP 10 months; SDHB [n = 4]: DCR 75%, SDP 10 months)., Conclusions: We demonstrate in a real-life clinical setting that all current therapies show reasonable efficacy in preventing disease progression, and this is equally true for patients with germline SDHB mutations., Competing Interests: Conflict of interest: None declared., (© The Author(s) 2023. Published by Oxford University Press on behalf of European Society of Endocrinology.)
- Published
- 2023
- Full Text
- View/download PDF
11. Metastatic Pheochromocytoma and Paraganglioma: Somatostatin Receptor 2 Expression, Genetics, and Therapeutic Responses.
- Author
-
Fischer A, Kloos S, Maccio U, Friemel J, Remde H, Fassnacht M, Pamporaki C, Eisenhofer G, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Zitzmann K, Bechmann N, Žygienė G, Richter S, Hantel C, Vetter D, Lehmann K, Mohr H, Pellegata NS, Ullrich M, Pietzsch J, Ziegler CG, Bornstein SR, Kroiss M, Reincke M, Pacak K, Grossman AB, Beuschlein F, and Nölting S
- Subjects
- Humans, Receptors, Somatostatin genetics, Receptors, Somatostatin metabolism, Retrospective Studies, Succinate Dehydrogenase genetics, Succinate Dehydrogenase metabolism, Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms therapy, Adrenal Gland Neoplasms metabolism, Neoplasms, Second Primary, Paraganglioma genetics, Paraganglioma therapy, Paraganglioma metabolism, Pheochromocytoma genetics, Pheochromocytoma therapy, Pheochromocytoma metabolism
- Abstract
Context: Pheochromocytomas and paragangliomas (PPGLs) with pathogenic mutations in the succinate dehydrogenase subunit B (SDHB) are associated with a high metastatic risk. Somatostatin receptor 2 (SSTR2)-dependent imaging is the most sensitive imaging modality for SDHB-related PPGLs, suggesting that SSTR2 expression is a significant cell surface therapeutic biomarker of such tumors., Objective: Exploration of the relationship between SSTR2 immunoreactivity and SDHB immunoreactivity, mutational status, and clinical behavior of PPGLs. Evaluation of SSTR-based therapies in metastatic PPGLs., Methods: Retrospective analysis of a multicenter cohort of PPGLs at 6 specialized Endocrine Tumor Centers in Germany, The Netherlands, and Switzerland. Patients with PPGLs participating in the ENSAT registry were included. Clinical data were extracted from medical records, and immunohistochemistry (IHC) for SDHB and SSTR2 was performed in patients with available tumor tissue. Immunoreactivity of SSTR2 was investigated using Volante scores. The main outcome measure was the association of SSTR2 IHC positivity with genetic and clinical-pathological features of PPGLs., Results: Of 202 patients with PPGLs, 50% were SSTR2 positive. SSTR2 positivity was significantly associated with SDHB- and SDHx-related PPGLs, with the strongest SSTR2 staining intensity in SDHB-related PPGLs (P = .01). Moreover, SSTR2 expression was significantly associated with metastatic disease independent of SDHB/SDHx mutation status (P < .001). In metastatic PPGLs, the disease control rate with first-line SSTR-based radionuclide therapy was 67% (n = 22, n = 11 SDHx), and with first-line "cold" somatostatin analogs 100% (n = 6, n = 3 SDHx)., Conclusion: SSTR2 expression was independently associated with SDHB/SDHx mutations and metastatic disease. We confirm a high disease control rate of somatostatin receptor-based therapies in metastatic PPGLs., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2023
- Full Text
- View/download PDF
12. Simultaneous magnetic resonance imaging of pH, perfusion and renal filtration using hyperpolarized 13 C-labelled Z-OMPD.
- Author
-
Grashei M, Wodtke P, Skinner JG, Sühnel S, Setzer N, Metzler T, Gulde S, Park M, Witt D, Mohr H, Hundshammer C, Strittmatter N, Pellegata NS, Steiger K, and Schilling F
- Subjects
- Animals, Rats, Perfusion, Glomerular Filtration Rate, Hydrogen-Ion Concentration, Magnetic Resonance Imaging, Filtration
- Abstract
pH alterations are a hallmark of many pathologies including cancer and kidney disease. Here, we introduce [1,5-
13 C2 ]Z-OMPD as a hyperpolarized extracellular pH and perfusion sensor for MRI which allows to generate a multiparametric fingerprint of renal disease status and to detect local tumor acidification. Exceptional long T1 of two minutes at 1 T, high pH sensitivity of up to 1.9 ppm per pH unit and suitability of using the C1 -label as internal frequency reference enables pH imaging in vivo of three pH compartments in healthy rat kidneys. Spectrally selective targeting of both13 C-resonances enables simultaneous imaging of perfusion and filtration in 3D and pH in 2D within one minute to quantify renal blood flow, glomerular filtration rates and renal pH in healthy and hydronephrotic kidneys with superior sensitivity compared to clinical routine methods. Imaging multiple biomarkers within a single session renders [1,5-13 C2 ]Z-OMPD a promising new hyperpolarized agent for oncology and nephrology., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
13. Editorial: New insights into multiple endocrine neoplasia type 1.
- Author
-
Barlier A, Romanet P, and Pellegata NS
- Subjects
- Humans, Multiple Endocrine Neoplasia Type 1 genetics
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
14. Identification of a Novel SSTR3 Full Agonist for the Treatment of Nonfunctioning Pituitary Adenomas.
- Author
-
Modena D, Moras ML, Sandrone G, Stevenazzi A, Vergani B, Dasgupta P, Kliever A, Gulde S, Marangelo A, Schillmaier M, Luque RM, Bäuerle S, Pellegata NS, Schulz S, and Steinkühler C
- Abstract
Somatostatin receptor (SSTR) agonists have been extensively used for treating neuroendocrine tumors. Synthetic therapeutic agonists showing selectivity for SSTR2 (Octreotide) or for SSTR2 and SSTR5 (Pasireotide) have been approved for the treatment of patients with acromegaly and Cushing's syndrome, as their pituitary tumors highly express SSTR2 or SSTR2/SSTR5, respectively. Nonfunctioning pituitary adenomas (NFPAs), which express high levels of SSTR3 and show only modest response to currently available SSTR agonists, are often invasive and cannot be completely resected, and therefore easily recur. The aim of the present study was the evaluation of ITF2984, a somatostatin analog and full SSTR3 agonist, as a new potential treatment for NFPAs. ITF2984 shows a 10-fold improved affinity for SSTR3 compared to Octreotide or Pasireotide. Molecular modeling and NMR studies indicated that the higher affinity for SSTR3 correlates with a higher stability of a distorted β-I turn in the cyclic peptide backbone. ITF2984 induces receptor internalization and phosphorylation, and triggers G-protein signaling at pharmacologically relevant concentrations. Furthermore, ITF2984 displays antitumor activity that is dependent on SSTR3 expression levels in the MENX (homozygous mutant) NFPA rat model, which closely recapitulates human disease. Therefore, ITF2984 may represent a novel therapeutic option for patients affected by NFPA.
- Published
- 2023
- Full Text
- View/download PDF
15. Inceptor correlates with markers of prostate cancer progression and modulates insulin/IGF1 signaling and cancer cell migration.
- Author
-
Wissmiller K, Bilekova S, Franko A, Lutz SZ, Katsburg M, Gulde S, Pellegata NS, Stenzl A, Heni M, Berti L, Häring HU, and Lickert H
- Subjects
- Male, Humans, Insulin-Like Growth Factor I metabolism, Prostate metabolism, Androgens, Androgen Antagonists, Cell Movement, Prostatic Neoplasms metabolism, Insulins
- Abstract
Objective: The insulin/insulin-like growth factor 1 (IGF1) pathway is emerging as a crucial component of prostate cancer progression. Therefore, we investigated the role of the novel insulin/IGF1 signaling modulator inceptor in prostate cancer., Methods: We analyzed the expression of inceptor in human samples of benign prostate epithelium and prostate cancer. Further, we performed signaling and functional assays using prostate cancer cell lines., Results: We found that inceptor was expressed in human benign and malignant prostate tissue and its expression positively correlated with various genes of interest, including genes involved in androgen signaling. In vitro, total levels of inceptor were increased upon androgen deprivation and correlated with high levels of androgen receptor in the nucleus. Inceptor overexpression was associated with increased cell migration, altered IGF1R trafficking and higher IGF1R activation., Conclusions: Our in vitro results showed that inceptor expression was associated with androgen status, increased migration, and IGF1R signaling. In human samples, inceptor expression was significantly correlated with markers of prostate cancer progression. Taken together, these data provide a basis for investigation of inceptor in the context of prostate cancer., (Copyright © 2023 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Combined Targeting of Pathogenetic Mechanisms in Pancreatic Neuroendocrine Tumors Elicits Synergistic Antitumor Effects.
- Author
-
Gulde S, Foscarini A, April-Monn SL, Genio E, Marangelo A, Satam S, Helbling D, Falconi M, Toledo RA, Schrader J, Perren A, Marinoni I, and Pellegata NS
- Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are the second most common malignancy of the pancreas. Surgery remains the only curative treatment for localized disease. For patients with inoperable advanced or metastatic disease, few targeted therapies are available, but their efficacy is unpredictable and variable. Exploiting prior knowledge on pathogenetic processes involved in PanNEN tumorigenesis, we tested buparlisib (PI3K inhibitor) and ribociclib (CDK4/6 inhibitor), as single agents or in combination, in different preclinical models. First, we used cell lines representative of well-differentiated (INS-1E, NT-3) and poorly differentiated (BON-1) PanNENs. The combination of buparlisib with ribociclib reduced the proliferation of 2D and 3D spheroid cultures more potently than the individual drugs. Buparlisib, but not ribociclib, induced apoptosis. The anti-proliferative activity of the drugs correlated with downstream target inhibition at mRNA and protein levels. We then tested the drugs on primary islet microtissues from a genetic PanNET animal model ( Men1 -defective mice) and from wild-type mice: the drug combination was effective against the former without altering islet cell physiology. Finally, we treated PanNET patient-derived islet-like 3D tumoroids: the combination of buparlisib with ribociclib was effective in three out of four samples. Combined targeting of PI3K and CDK4/6 is a promising strategy for PanNENs spanning various molecular and histo-pathological features.
- Published
- 2022
- Full Text
- View/download PDF
17. Obesity and cancer-extracellular matrix, angiogenesis, and adrenergic signaling as unusual suspects linking the two diseases.
- Author
-
Pellegata NS, Berriel Diaz M, Rohm M, and Herzig S
- Subjects
- Adipose Tissue, Extracellular Matrix, Humans, Obesity complications, Adrenergic Agents, Neoplasms epidemiology
- Abstract
Obesity is an established risk factor for several human cancers. Given the association between excess body weight and cancer, the increasing rates of obesity worldwide are worrisome. A variety of obesity-related factors has been implicated in cancer initiation, progression, and response to therapy. These factors include circulating nutritional factors, hormones, and cytokines, causing hyperinsulinemia, inflammation, and adipose tissue dysfunction. The impact of these conditions on cancer development and progression has been the focus of extensive literature. In this review, we concentrate on processes that can link obesity and cancer, and which provide a novel perspective: extracellular matrix remodeling, angiogenesis, and adrenergic signaling. We describe molecular mechanisms involved in these processes, which represent putative targets for intervention. Liver, pancreas, and breast cancers were chosen as exemplary disease models. In view of the expanding epidemic of obesity, a better understanding of the tumorigenic process in obese individuals might lead to more effective treatments and preventive measures., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. Angpt2/Tie2 autostimulatory loop controls tumorigenesis.
- Author
-
Karabid NM, Wiedemann T, Gulde S, Mohr H, Segaran RC, Geppert J, Rohm M, Vitale G, Gaudenzi G, Dicitore A, Ankerst DP, Chen Y, Braren R, Kaissis G, Schilling F, Schillmaier M, Eisenhofer G, Herzig S, Roncaroli F, Honegger JB, and Pellegata NS
- Subjects
- Animals, Carcinogenesis, Endothelial Cells metabolism, Heterografts, Humans, Mice, Neoplasm Recurrence, Local, Rats, Receptor, TIE-2 genetics, Receptor, TIE-2 metabolism, Zebrafish, Angiopoietin-2 metabolism, Pituitary Neoplasms genetics, Pituitary Neoplasms metabolism, Pituitary Neoplasms pathology
- Abstract
Invasive nonfunctioning (NF) pituitary neuroendocrine tumors (PitNETs) are non-resectable neoplasms associated with frequent relapses and significant comorbidities. As the current therapies of NF-PitNETs often fail, new therapeutic targets are needed. The observation that circulating angiopoietin-2 (ANGPT2) is elevated in patients with NF-PitNET and correlates with tumor aggressiveness prompted us to investigate the ANGPT2/TIE2 axis in NF-PitNETs in the GH3 PitNET cell line, primary human NF-PitNET cells, xenografts in zebrafish and mice, and in MENX rats, the only autochthonous NF-PitNET model. We show that PitNET cells express a functional TIE2 receptor and secrete bioactive ANGPT2, which promotes, besides angiogenesis, tumor cell growth in an autocrine and paracrine fashion. ANGPT2 stimulation of TIE2 in tumor cells activates downstream cell proliferation signals, as previously demonstrated in endothelial cells (ECs). Tie2 gene deletion blunts PitNETs growth in xenograft models, and pharmacological inhibition of Angpt2/Tie2 signaling antagonizes PitNETs in primary cell cultures, tumor xenografts in mice, and in MENX rats. Thus, the ANGPT2/TIE2 axis provides an exploitable therapeutic target in NF-PitNETs and possibly in other tumors expressing ANGPT2/TIE2. The ability of tumor cells to coopt angiogenic signals classically viewed as EC-specific expands our view on the microenvironmental cues that are essential for tumor progression., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
19. Personalized drug testing in human pheochromocytoma/paraganglioma primary cultures.
- Author
-
Wang K, Schütze I, Gulde S, Bechmann N, Richter S, Helm J, Lauseker M, Maurer J, Reul A, Spoettl G, Klink B, William D, Knösel T, Friemel J, Bihl M, Weber A, Fankhauser M, Schober L, Vetter D, Broglie Däppen M, Ziegler CG, Ullrich M, Pietzsch J, Bornstein SR, Lottspeich C, Kroiss M, Fassnacht M, Wenter VUJ, Ladurner R, Hantel C, Reincke M, Eisenhofer G, Grossman AB, Pacak K, Beuschlein F, Auernhammer CJ, Pellegata NS, and Nölting S
- Subjects
- Animals, Everolimus therapeutic use, Humans, Mice, Zoledronic Acid therapeutic use, Adrenal Gland Neoplasms drug therapy, Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Paraganglioma drug therapy, Paraganglioma genetics, Paraganglioma pathology, Pheochromocytoma drug therapy, Pheochromocytoma genetics, Pheochromocytoma metabolism
- Abstract
Aggressive pheochromocytomas and paragangliomas (PPGLs) are difficult to treat, and molecular targeting is being increasingly considered, but with variable results. This study investigates established and novel molecular-targeted drugs and chemotherapeutic agents for the treatment of PPGLs in human primary cultures and murine cell line spheroids. In PPGLs from 33 patients, including 7 metastatic PPGLs, we identified germline or somatic driver mutations in 79% of cases, allowing us to assess potential differences in drug responsivity between pseudohypoxia-associated cluster 1-related (n = 10) and kinase signaling-associated cluster 2-related (n = 14) PPGL primary cultures. Single anti-cancer drugs were either more effective in cluster 1 (cabozantinib, selpercatinib, and 5-FU) or similarly effective in both clusters (everolimus, sunitinib, alpelisib, trametinib, niraparib, entinostat, gemcitabine, AR-A014418, and high-dose zoledronic acid). High-dose estrogen and low-dose zoledronic acid were the only single substances more effective in cluster 2. Neither cluster 1- nor cluster 2-related patient primary cultures responded to HIF-2a inhibitors, temozolomide, dabrafenib, or octreotide. We showed particular efficacy of targeted combination treatments (cabozantinib/everolimus, alpelisib/everolimus, alpelisib/trametinib) in both clusters, with higher efficacy of some targeted combinations in cluster 2 and overall synergistic effects (cabozantinib/everolimus, alpelisib/trametinib) or synergistic effects in cluster 2 (alpelisib/everolimus). Cabozantinib/everolimus combination therapy, gemcitabine, and high-dose zoledronic acid appear to be promising treatment options with particularly high efficacy in SDHB-mutant and metastatic tumors. In conclusion, only minor differences regarding drug responsivity were found between cluster 1 and cluster 2: some single anti-cancer drugs were more effective in cluster 1 and some targeted combination treatments were more effective in cluster 2.
- Published
- 2022
- Full Text
- View/download PDF
20. Adipocyte-specific tribbles pseudokinase 1 regulates plasma adiponectin and plasma lipids in mice.
- Author
-
Ha EE, Quartuccia GI, Ling R, Xue C, Karikari RA, Hernandez-Ono A, Hu KY, Matias CV, Imam R, Cui J, Pellegata NS, Herzig S, Georgiadi A, Soni RK, and Bauer RC
- Subjects
- Adipocytes metabolism, Animals, Intracellular Signaling Peptides and Proteins, Mice, Mice, Knockout, Protein Serine-Threonine Kinases antagonists & inhibitors, Triglycerides metabolism, Adiponectin genetics, Adiponectin metabolism, Genome-Wide Association Study
- Abstract
Objective: Multiple genome-wide association studies (GWAS) have identified SNPs in the 8q24 locus near TRIB1 that are significantly associated with plasma lipids and other markers of cardiometabolic health, and prior studies have revealed the roles of hepatic and myeloid Trib1 in plasma lipid regulation and atherosclerosis. The same 8q24 SNPs are additionally associated with plasma adiponectin levels in humans, implicating TRIB1 in adipocyte biology. Here, we hypothesize that TRIB1 in adipose tissue regulates plasma adiponectin, lipids, and metabolic health., Methods: We investigate the metabolic phenotype of adipocyte-specific Trib1 knockout mice (Trib1_ASKO) fed on chow and high-fat diet (HFD). Through secretomics of adipose tissue explants and RNA-seq of adipocytes and livers from these mice, we further investigate the mechanism of TRIB1 in adipose tissue., Results: Trib1_ASKO mice have an improved metabolic phenotype with increased plasma adiponectin levels, improved glucose tolerance, and decreased plasma lipids. Trib1_ASKO adipocytes have increased adiponectin production and secretion independent of the known TRIB1 function of regulating proteasomal degradation. RNA-seq analysis of adipocytes and livers from Trib1_ASKO mice indicates that alterations in adipocyte function underlie the observed plasma lipid changes. Adipose tissue explant secretomics further reveals that Trib1_ASKO adipose tissue has decreased ANGPTL4 production, and we demonstrate an accompanying increase in the lipoprotein lipase (LPL) activity that likely underlies the triglyceride phenotype., Conclusions: This study shows that adipocyte Trib1 regulates multiple aspects of metabolic health, confirming previously observed genetic associations in humans and shedding light on the further mechanisms by which TRIB1 regulates plasma lipids and metabolic health., (Copyright © 2021 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. Imaging pheochromocytoma in small animals: preclinical models to improve diagnosis and treatment.
- Author
-
Mohr H, Foscarini A, Steiger K, Ballke S, Rischpler C, Schilling F, and Pellegata NS
- Abstract
Pheochromocytomas (PCCs) and paragangliomas (PGLs), together referred to as PPGLs, are rare chromaffin cell-derived tumors. They require timely diagnosis as this is the only way to achieve a cure through surgery and because of the potentially serious cardiovascular complications and sometimes life-threatening comorbidities that can occur if left untreated. The biochemical diagnosis of PPGLs has improved over the last decades, and the knowledge of the underlying genetics has dramatically increased. In addition to conventional anatomical imaging by CT and MRI for PPGL detection, new functional imaging modalities have emerged as very useful for patient surveillance and stratification for therapy. The availability of validated and predictive animal models of cancer is essential for translating molecular, imaging and therapy response findings from the bench to the bedside. This is especially true for rare tumors, such as PPGLs, for which access to large cohorts of patients is limited. There are few animal models of PPGLs that have been instrumental in refining imaging modalities for early tumor detection, as well as in identifying and evaluating novel imaging tracers holding promise for the detection and/or treatment of human PPGLs. The in vivo PPGL models mainly include xenografts/allografts generated by engrafting rat or mouse cell lines, as no representative human cell line is available. In addition, there is a model of endogenous PCCs (i.e., MENX rats) that was characterized in our laboratory. In this review, we will summarize the contribution that various representative models of PPGL have given to the visualization of these tumors in vivo and we present an example of a tracer first evaluated in MENX rats, and then translated to the detection of these tumors in human patients. In addition, we will illustrate briefly the potential of ex vivo biological imaging of intact adrenal glands in MENX rats., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. Editorial: Stem Cells in Endocrine Tumors.
- Author
-
Di Franco S, Pellegata NS, Luconi M, and Stassi G
- Subjects
- Adult, Animals, Biomarkers, Tumor physiology, Cell Proliferation, Disease Progression, Endocrine Gland Neoplasms diagnosis, Endocrine Gland Neoplasms metabolism, Energy Metabolism physiology, Epithelial-Mesenchymal Transition, Female, Humans, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Pregnancy, Endocrine Gland Neoplasms pathology, Neoplastic Stem Cells physiology
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
23. Gender-Specific Efficacy Revealed by Head-to-Head Comparison of Pasireotide and Octreotide in a Representative In Vivo Model of Nonfunctioning Pituitary Tumors.
- Author
-
Gulde S, Wiedemann T, Schillmaier M, Valença I, Lupp A, Steiger K, Yen HY, Bäuerle S, Notni J, Luque R, Schmid H, Schulz S, Ankerst DP, Schilling F, and Pellegata NS
- Abstract
Invasive nonfunctioning pituitary tumors (NFPTs) are non-resectable neoplasms associated with frequent relapse and significant comorbidities. Current treatments, including somatostatin receptor 2 (SSTR2)-directed somatostatin analogs (SSAs), often fail against NFPTs. Thus, identifying effective therapies is clinically relevant. As NFPTs express SSTR3 at high levels, pasireotide, a multireceptor-targeted SSA, might be beneficial. Here we evaluated pasireotide in the only representative model of spontaneous NFPTs (MENX rats) in vivo. Octreotide long-acting release (LAR), pasireotide LAR, or placebo, were administered to age-matched, tumor-bearing MENX rats of both sexes for 28 d or 56 d. Longitudinal high-resolution magnetic resonance imaging monitored tumor growth. While tumors in placebo-treated rats increased in volume over time, PTs in drug-treated rats displayed significant growth suppression, and occasional tumor shrinkage. Pasireotide elicited stronger growth inhibition. Radiological responses correlated with tumors' proliferation rates. Both SSAs, but especially pasireotide, were more effective in female vs. male rats. Basal Sstr3 expression was significantly higher in the former group. It is noteworthy that female human NFPTs patients also have a trend towards higher SSTR3 expression. Altogether, our studies provide the rationale for testing pasireotide in patients with residual/recurrent NFPTs. If confirmed, the sex-related SSTR3 expression might be used as criteria to stratify NFPTs patients for treatment with pasireotide.
- Published
- 2021
- Full Text
- View/download PDF
24. Mutation of the Cell Cycle Regulator p27kip1 Drives Pseudohypoxic Pheochromocytoma Development.
- Author
-
Mohr H, Ballke S, Bechmann N, Gulde S, Malekzadeh-Najafabadi J, Peitzsch M, Ntziachristos V, Steiger K, Wiedemann T, and Pellegata NS
- Abstract
Background: Pseudohypoxic tumors activate pro-oncogenic pathways typically associated with severe hypoxia even when sufficient oxygen is present, leading to highly aggressive tumors. Prime examples are pseudohypoxic pheochromocytomas and paragangliomas (p-PPGLs), neuroendendocrine tumors currently lacking effective therapy. Previous attempts to generate mouse models for p-PPGLs all failed. Here, we describe that the rat MENX line, carrying a Cdkn1b (p27) frameshift-mutation, spontaneously develops pseudohypoxic pheochromocytoma (p-PCC)., Methods: We compared rat p-PCCs with their cognate human tumors at different levels: histology, immunohistochemistry, catecholamine profiling, electron microscopy, transcriptome and metabolome. The vessel architecture and angiogenic potential of pheochromocytomas (PCCs) was analyzed by light-sheet fluorescence microscopy ex vivo and multi-spectral optoacoustic tomography (MSOT) in vivo., Results: The analysis of tissues at various stages, from hyperplasia to advanced grades, allowed us to correlate tumor characteristics with progression. Pathological changes affecting the mitochrondrial ultrastructure where present already in hyperplasias. Rat PCCs secreted high levels of norepinephrine and dopamine. Transcriptomic and metabolomic analysis revealed changes in oxidative phosphorylation that aggravated over time, leading to an accumulation of the oncometabolite 2-hydroxyglutarate, and to hypermethylation, evident by the loss of the epigenetic mark 5-hmC. While rat PCC xenografts showed high oxygenation, induced by massive neoangiogenesis, rat primary PCC transcriptomes possessed a pseudohypoxic signature of high Hif2a , Vegfa , and low Pnmt expression, thereby clustering with human p-PPGL., Conclusion: Endogenous rat PCCs recapitulate key phenotypic features of human p-PPGLs. Thus, MENX rats emerge as the best available animal model of these aggressive tumors. Our study provides evidence of a link between cell cycle dysregulation and pseudohypoxia.
- Published
- 2021
- Full Text
- View/download PDF
25. miR-34a is upregulated in AIP-mutated somatotropinomas and promotes octreotide resistance.
- Author
-
Bogner EM, Daly AF, Gulde S, Karhu A, Irmler M, Beckers J, Mohr H, Beckers A, and Pellegata NS
- Subjects
- Animals, Cell Line, Cell Movement, Cell Proliferation, Female, Fibroblasts cytology, Fibroblasts metabolism, Gene Expression Regulation, Neoplastic, Germ-Line Mutation, Growth Hormone-Secreting Pituitary Adenoma drug therapy, Humans, Male, Mice, Octreotide pharmacology, Octreotide therapeutic use, Pituitary Neoplasms drug therapy, Drug Resistance, Neoplasm, Growth Hormone-Secreting Pituitary Adenoma genetics, Intracellular Signaling Peptides and Proteins genetics, MicroRNAs genetics, Pituitary Neoplasms genetics, Up-Regulation
- Abstract
Pituitary adenomas (PAs) are intracranial tumors associated with significant morbidity due to hormonal dysregulation, mass effects and have a heavy treatment burden. Growth hormone (GH)-secreting PAs (somatotropinomas) cause acromegaly-gigantism. Genetic forms of somatotropinomas due to germline AIP mutations (AIPmut+) have an early onset and are aggressive and resistant to treatment with somatostatin analogs (SSAs), including octreotide. The molecular underpinnings of these clinical features remain unclear. We investigated the role of miRNA dysregulation in AIPmut+ vs AIPmut- PA samples by array analysis. miR-34a and miR-145 were highly expressed in AIPmut+ vs AIPmut- somatotropinomas. Ectopic expression of AIPmut (p.R271W) in Aip
-/- mouse embryonic fibroblasts (MEFs) upregulated miR-34a and miR-145, establishing a causal link between AIPmut and miRNA expression. In PA cells (GH3), miR-34a overexpression promoted proliferation, clonogenicity, migration and suppressed apoptosis, whereas miR-145 moderately affected proliferation and apoptosis. Moreover, high miR-34a expression increased intracellular cAMP, a critical mitogenic factor in PAs. Crucially, high miR-34a expression significantly blunted octreotide-mediated GH inhibition and antiproliferative effects. miR-34a directly targets Gnai2 encoding Gαi2, a G protein subunit inhibiting cAMP production. Accordingly, Gαi2 levels were significantly lower in AIPmut+ vs AIPmut- PA. Taken together, somatotropinomas with AIP mutations overexpress miR-34a, which in turn downregulates Gαi2 expression, increases cAMP concentration and ultimately promotes cell growth. Upregulation of miR-34a also impairs the hormonal and antiproliferative response of PA cells to octreotide. Thus, miR-34a is a novel downstream target of mutant AIP that promotes a cellular phenotype mirroring the aggressive clinical features of AIPmut+ acromegaly., (© 2020 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of Union for International Cancer Control.)- Published
- 2020
- Full Text
- View/download PDF
26. AIP-mutated acromegaly resistant to first-generation somatostatin analogs: long-term control with pasireotide LAR in two patients.
- Author
-
Daly AF, Rostomyan L, Betea D, Bonneville JF, Villa C, Pellegata NS, Waser B, Reubi JC, Waeber Stephan C, Christ E, and Beckers A
- Abstract
Acromegaly is a rare disease due to chronic excess growth hormone (GH) and IGF-1. Aryl hydrocarbon receptor interacting protein (AIP) mutations are associated with an aggressive, inheritable form of acromegaly that responds poorly to SST2-specific somatostatin analogs (SSA). The role of pasireotide, an SSA with affinity for multiple SSTs, in patients with AIP mutations has not been reported. We studied two AIP mutation positive acromegaly patients with early-onset, invasive macroadenomas and inoperable residues after neurosurgery. Patient 1 came from a FIPA kindred and had uncontrolled GH/IGF-1 throughout 10 years of octreotide/lanreotide treatment. When switched to pasireotide LAR, he rapidly experienced hormonal control which was associated with marked regression of his tumor residue. Pasireotide LAR was stopped after >10 years due to low IGF-1 and he maintained hormonal control without tumor regrowth for >18 months off pasireotide LAR. Patient 2 had a pituitary adenoma diagnosed when aged 17 that was not cured by surgery. Chronic pasireotide LAR therapy produced hormonal control and marked tumor shrinkage but control was lost when switched to octreotide. Tumor immunohistochemistry showed absent AIP and SST2 staining and positive SST5. Her AIP mutation positive sister developed a 2.5 cm follicular thyroid carcinoma aged 21 with tumoral loss of heterozygosity at the AIP locus and absent AIP staining. Patients 1 and 2 required multi-modal therapy to control diabetes. On stopping pasireotide LAR after >10 years of treatment, Patient 1's glucose metabolism returned to baseline levels. Long-term pasireotide LAR therapy can be beneficial in some AIP mutation positive acromegaly patients that are resistant to first-generation SSA.
- Published
- 2019
- Full Text
- View/download PDF
27. Adiponectin affects the mechanical responses in strips from the mouse gastric fundus.
- Author
-
Idrizaj E, Garella R, Castellini G, Mohr H, Pellegata NS, Francini F, Ricca V, Squecco R, and Baccari MC
- Subjects
- Adipocytes metabolism, Adipose Tissue, White cytology, Adipose Tissue, White metabolism, Animals, Electric Stimulation, Female, Mice, Mice, Inbred C57BL, Muscle Contraction physiology, Muscle Relaxation physiology, Receptors, Adiponectin metabolism, Adiponectin physiology, Gastric Fundus physiology, Muscle, Smooth physiology
- Abstract
Aim: To investigate whether the adipocytes derived hormone adiponectin (ADPN) affects the mechanical responses in strips from the mouse gastric fundus., Methods: For functional experiments, gastric strips from the fundal region were cut in the direction of the longitudinal muscle layer and placed in organ baths containing Krebs-Henseleit solution. Mechanical responses were recorded via force-displacement transducers, which were coupled to a polygraph for continuous recording of isometric tension. Electrical field stimulation (EFS) was applied via two platinum wire rings through which the preparation was threaded. The effects of ADPN were investigated on the neurally-induced contractile and relaxant responses elicited by EFS. The expression of ADPN receptors, Adipo-R1 and Adipo-R2, was also evaluated by touchdown-PCR analysis., Results: In the functional experiments, EFS (4-16 Hz) elicited tetrodotoxin (TTX)-sensitive contractile responses. Addition of ADPN to the bath medium caused a reduction in amplitude of the neurally-induced contractile responses ( P < 0.05). The effects of ADPN were no longer observed in the presence of the nitric oxide (NO) synthesis inhibitor L-N
G -nitro arginine (L-NNA) ( P > 0.05). The direct smooth muscle response to methacholine was not influenced by ADPN ( P > 0.05). In carbachol precontracted strips and in the presence of guanethidine, EFS induced relaxant responses. Addition of ADPN to the bath medium, other than causing a slight and progressive decay of the basal tension, increased the amplitude of the neurally-induced relaxant responses ( P < 0.05). Touchdown-PCR analysis revealed the expression of both Adipo-R1 and Adipo-R2 in the gastric fundus., Conclusion: The results indicate for the first time that ADPN is able to influence the mechanical responses in strips from the mouse gastric fundus., Competing Interests: Conflict-of-interest statement: No conflicts of interest, financial or otherwise, are declared by the authors.- Published
- 2018
- Full Text
- View/download PDF
28. Characterization of neuroendocrine tumors in heterozygous mutant MENX rats: a novel model of invasive medullary thyroid carcinoma.
- Author
-
Molatore S, Kügler A, Irmler M, Wiedemann T, Neff F, Feuchtinger A, Beckers J, Robledo M, Roncaroli F, and Pellegata NS
- Subjects
- Animals, Cyclin-Dependent Kinase Inhibitor p27 genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Male, Rats, Rats, Mutant Strains, Transcriptome, Adrenal Gland Neoplasms genetics, Carcinoma, Neuroendocrine genetics, Disease Models, Animal, Pheochromocytoma genetics, Pituitary Neoplasms genetics, Thyroid Neoplasms genetics
- Abstract
Rats affected by the MENX syndrome spontaneously develop multiple neuroendocrine tumors (NETs) including adrenal, pituitary and thyroid gland neoplasms. MENX was initially reported to be inherited as a recessive trait and affected rats were found to be homozygous for the predisposing Cdkn1b mutation encoding p27. We here report that heterozygous MENX-mutant rats (p27+/mut) develop the same spectrum of NETs seen in the homozygous (p27mut/mut) animals but with slower progression. Consequently, p27+/mut rats have a significantly shorter lifespan compared with their wild-type (p27+/+) littermates. In the tumors of p27+/mut rats, the wild-type Cdkn1b allele is neither lost nor silenced, implying that p27 is haploinsufficient for tumor suppression in this model. Transcriptome profiling of rat adrenal (pheochromocytoma) and pituitary tumors having different p27 dosages revealed a tissue-specific, dose-dependent effect of p27 on gene expression. In p27+/mut rats, thyroid neoplasms progress to invasive and metastatic medullary thyroid carcinomas (MTCs) accompanied by increased calcitonin levels, as in humans. Comparison of expression signatures of late-stage vs early-stage MTCs from p27+/mut rats identified genes potentially involved in tumor aggressiveness. The expression of a subset of these genes was evaluated in human MTCs and found to be associated with aggressive RET-M918T-positive tumors. Altogether, p27 haploinsufficiency in MENX rats uncovered a novel, representative model of invasive and metastatic MTC exploitable for translational studies of this often aggressive and incurable cancer., (© 2018 Society for Endocrinology.)
- Published
- 2018
- Full Text
- View/download PDF
29. Animal models of MEN1.
- Author
-
Mohr H and Pellegata NS
- Subjects
- Animals, Disease Models, Animal, Drosophila, Fishes, Mice, Mice, Knockout, Multiple Endocrine Neoplasia Type 1 metabolism, Multiple Endocrine Neoplasia Type 1 pathology, Rats, Drosophila Proteins metabolism, Multiple Endocrine Neoplasia Type 1 genetics
- Abstract
Animal models of cancer have been instrumental in advancing our understanding of the biology of tumor initiation and progression, in studying gene function and in performing preclinical studies aimed at testing novel therapies. Several animal models of the MEN1 syndrome have been generated in different organisms by introducing loss-of-function mutations in the orthologues of the human MEN1 gene. In this review, we will discuss MEN1 and MEN1-like models in Drosophila, mice and rats. These model systems with their specific advantages and limitations have contributed to elucidate the function of Menin in tumorigenesis, which turned out to be remarkably conserved from flies to mammals, as well as the biology of the disease. Mouse models of MEN1 closely resemble the human disease in terms of tumor spectrum and associated hormonal changes, although individual tumor frequencies are variable. Rats affected by the MENX (MEN1-like) syndrome share some features with MEN1 patients albeit they bear a germline mutation in Cdkn1b (p27) and not in Men1 Both Men1 -knockout mice and MENX rats have been exploited for therapy-response studies testing novel drugs for efficacy against neuroendocrine tumors (NETs) and have provided promising leads for novel therapies. In addition to presenting well-established models of MEN1, we also discuss potential models which, if implemented, might broaden even further our knowledge of neuroendocrine tumorigenesis. In the future, patient-derived xenografts in zebrafish or mice might allow us to expand the tool-box currently available for preclinical studies of MEN1-associated tumors., (© 2017 Society for Endocrinology.)
- Published
- 2017
- Full Text
- View/download PDF
30. Loss of p27 expression is associated with MEN1 gene mutations in sporadic parathyroid adenomas.
- Author
-
Borsari S, Pardi E, Pellegata NS, Lee M, Saponaro F, Torregrossa L, Basolo F, Paltrinieri E, Zatelli MC, Materazzi G, Miccoli P, Marcocci C, and Cetani F
- Subjects
- Adenoma metabolism, Adenoma pathology, Adult, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Female, Humans, Hyperparathyroidism, Primary metabolism, Hyperparathyroidism, Primary pathology, Immunohistochemistry, Male, Middle Aged, Parathyroid Neoplasms pathology, Proto-Oncogene Proteins metabolism, Adenoma genetics, Cyclin-Dependent Kinase Inhibitor p27 genetics, Hyperparathyroidism, Primary genetics, Mutation, Parathyroid Neoplasms genetics, Proto-Oncogene Proteins genetics
- Abstract
MEN1 is the main gene responsible for tumorigenesis of syndromic and sporadic primary hyperparathyroidism (PHPT). Germline mutations of the CDKN1B/p27
Kip gene have been associated with multiple endocrine tumors in rats and humans. To evaluate the involvement of the CDKN1B gene and its relationship with MEN1 in sporadic PHPT, we carried out sequencing and loss of heterozygosity analyses of the CDKN1B gene in 147 sporadic parathyroid adenomas. p27 immunohistochemistry and genetic screening of the MEN1 gene were performed in 50 cases. Three germline CDKN1B variants (c.-80C>T, c.-29_-26delAGAG, c.397C>A) were identified in 3/147 patients. Reduction of CDKN1B gene transcription rate was demonstrated in vitro for the novel c.-80C>T and the c.-29_-26delAGAG variants. Loss of p27 expression was detected in the tumor carrying the c.-29_-26delAGAG variant. Two tumors carrying the CDKN1B variants also harbored a MEN1 mutation. Fifty-four percent of 50 CDKN1B mutation-negative tumors had a reduction of p27 nuclear staining. Somatic MEN1 mutations, identified in 15/50 samples, significantly segregated in tumors negative for nuclear and cytoplasmic p27 staining. The germline nature of the CDKN1B mutations suggests that they might predispose to PHPT. The lack of somatic CDKN1B mutations in our samples points to a rare involvement in parathyroid adenomas, despite the frequent loss of nuclear p27 expression. MEN1 biallelic inactivation seems to be directly related to down-regulation of p27 expression through the inhibition of CDKN1B gene transcription.- Published
- 2017
- Full Text
- View/download PDF
31. Targeting PI3K/mTOR signaling exerts potent antitumor activity in pheochromocytoma in vivo.
- Author
-
Lee M, Minaskan N, Wiedemann T, Irmler M, Beckers J, Yousefi BH, Kaissis G, Braren R, Laitinen I, and Pellegata NS
- Subjects
- Adrenal Gland Neoplasms genetics, Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Imidazoles pharmacology, Norepinephrine Plasma Membrane Transport Proteins genetics, Pheochromocytoma genetics, Quinolines pharmacology, Rats, Mutant Strains, Rats, Sprague-Dawley, Signal Transduction, Adrenal Gland Neoplasms drug therapy, Antineoplastic Agents therapeutic use, Imidazoles therapeutic use, Pheochromocytoma drug therapy, Phosphoinositide-3 Kinase Inhibitors, Quinolines therapeutic use, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Pheochromocytomas (PCCs) are mostly benign tumors, amenable to complete surgical resection. However, 10-17% of cases can become malignant, and once metastasized, there is no curative treatment for this disease. Given the need to identify the effective therapeutic approaches for PCC, we evaluated the antitumor potential of the dual-PI3K/mTOR inhibitor BEZ235 against these tumors. We employed an in vivo model of endogenous PCCs (MENX mutant rats), which closely recapitulate the human tumors. Mutant rats with PCCs were treated with 2 doses of BEZ235 (20 and 30 mg/kg), or with placebo, for 2 weeks. Treatment with BEZ235 induced cytostatic and cytotoxic effects on rat PCCs, which could be appreciated by both staining the tumors ex vivo with appropriate markers and non-invasively by functional imaging (diffusion-weighted magnetic resonance imaging) in vivo Transcriptomic analyses of tumors from rats treated with BEZ235 or placebo-identified potential mediators of therapy response were performed. Slc6a2, encoding the norepinephrine transporter (NET), was downregulated in a dose-dependent manner by BEZ235 in rat PCCs. Moreover, BEZ235 reduced Slc6a2/NET expression in PCC cell lines (MPC) also. Studies of a BEZ235-resistant derivative of the MPC cell line confirmed that the reduction of NET expression associates with the response to the drug. Reduction of NET expression after BEZ235 treatment in vivo could be monitored by positron emission tomography (PET) using a tracer targeting NET. Altogether, here we demonstrate the efficacy of BEZ235 against PCC in vivo, and show that functional imaging can be employed to monitor the response of PCC to PI3K/mTOR inhibition therapy., (© 2017 Society for Endocrinology.)
- Published
- 2017
- Full Text
- View/download PDF
32. An AMP-activated protein kinase-stabilizing peptide ameliorates adipose tissue wasting in cancer cachexia in mice.
- Author
-
Rohm M, Schäfer M, Laurent V, Üstünel BE, Niopek K, Algire C, Hautzinger O, Sijmonsma TP, Zota A, Medrikova D, Pellegata NS, Ryden M, Kulyte A, Dahlman I, Arner P, Petrovic N, Cannon B, Amri EZ, Kemp BE, Steinberg GR, Janovska P, Kopecky J, Wolfrum C, Blüher M, Berriel Diaz M, and Herzig S
- Subjects
- AMP-Activated Protein Kinases pharmacology, Adipocytes, White metabolism, Adipose Tissue, White metabolism, Animals, Apoptosis Regulatory Proteins metabolism, Cachexia etiology, Cells, Cultured, In Vitro Techniques, Lipogenesis drug effects, Lipolysis drug effects, Mice, Neoplasms complications, Thermogenesis drug effects, Uncoupling Protein 1 drug effects, Uncoupling Protein 1 metabolism, AMP-Activated Protein Kinases metabolism, Adipocytes, White drug effects, Adipose Tissue, White drug effects, Apoptosis Regulatory Proteins drug effects, Cachexia metabolism, Lipid Metabolism drug effects, Neoplasms metabolism, Peptide Fragments pharmacology
- Abstract
Cachexia represents a fatal energy-wasting syndrome in a large number of patients with cancer that mostly results in a pathological loss of skeletal muscle and adipose tissue. Here we show that tumor cell exposure and tumor growth in mice triggered a futile energy-wasting cycle in cultured white adipocytes and white adipose tissue (WAT), respectively. Although uncoupling protein 1 (Ucp1)-dependent thermogenesis was dispensable for tumor-induced body wasting, WAT from cachectic mice and tumor-cell-supernatant-treated adipocytes were consistently characterized by the simultaneous induction of both lipolytic and lipogenic pathways. Paradoxically, this was accompanied by an inactivated AMP-activated protein kinase (Ampk), which is normally activated in peripheral tissues during states of low cellular energy. Ampk inactivation correlated with its degradation and with upregulation of the Ampk-interacting protein Cidea. Therefore, we developed an Ampk-stabilizing peptide, ACIP, which was able to ameliorate WAT wasting in vitro and in vivo by shielding the Cidea-targeted interaction surface on Ampk. Thus, our data establish the Ucp1-independent remodeling of adipocyte lipid homeostasis as a key event in tumor-induced WAT wasting, and we propose the ACIP-dependent preservation of Ampk integrity in the WAT as a concept in future therapies for cachexia.
- Published
- 2016
- Full Text
- View/download PDF
33. Primary Cell Culture Systems for Human Thyroid Studies.
- Author
-
Wang Y, Li W, Phay JE, Shen R, Pellegata NS, Saji M, Ringel MD, de la Chapelle A, and He H
- Subjects
- Cell Dedifferentiation physiology, Cells, Cultured, Humans, Primary Cell Culture methods, Thyroid Gland cytology
- Abstract
Background: Cell models are key instruments for in vitro studies of the thyroid. Permanent thyroid cell lines that are widely used in laboratory research typically originate from tumors. For many purposes, it is desirable to compare tumor cells with cells originating from normal tissue. However, such cultures grow slowly, have a highly limited life-span, and are known to lose their thyroid characteristics. The aim of the present study was to type coding and noncoding thyroid markers in different culture systems in an attempt to determine the optimal conditions for in vitro experimentation., Methods: Human primary thyroid cells were isolated from histologically non-tumorous tissues. Two alternative media (6H and h7H) were used. The morphology and behavior of the ensuing monolayer (two-dimensional) cultures was monitored by microscopy. The expression of key thyroid-related genes (n = 9) was monitored by reverse transcription polymerase chain reaction on days 8, 21, and 43 after initiation. As a pilot study, the same markers were studied in a three-dimensional hanging-drop culture system., Results: In the cultures with 6H or h7H medium, the primary thyroid cells displayed growth in numbers and size. Most cells retained the main morphological characteristics of thyroid cells throughout the first two weeks of culture, and fibroblast-like cells appeared around day 19. By day 21, most thyroid gene markers were retained, but by day 43, several markers were no longer present. The lncRNA transcripts PTCSC2 (spliced) and PTCSC3 were the first to disappear. There were no fundamental differences between the two media in the early period of culture. In the three-dimensional system, most thyroid markers were retained by day 21., Conclusion: Cultures of thyroid cells retain many thyroid characteristics up to day 21. Thereafter, fibroblast-like dedifferentiated cells begin to dominate.
- Published
- 2016
- Full Text
- View/download PDF
34. Morphology, Biochemistry, and Pathophysiology of MENX-Related Pheochromocytoma Recapitulate the Clinical Features.
- Author
-
Wiedemann T, Peitzsch M, Qin N, Neff F, Ehrhart-Bornstein M, Eisenhofer G, and Pellegata NS
- Subjects
- Adrenal Gland Neoplasms physiopathology, Adrenal Medulla metabolism, Adrenal Medulla pathology, Animals, Blood Pressure physiology, Disease Models, Animal, Epinephrine blood, Female, Male, Metanephrine blood, Multiple Endocrine Neoplasia physiopathology, Norepinephrine blood, Normetanephrine blood, Pheochromocytoma physiopathology, Rats, Rats, Mutant Strains, Rats, Sprague-Dawley, Adrenal Gland Neoplasms metabolism, Adrenal Gland Neoplasms pathology, Multiple Endocrine Neoplasia metabolism, Multiple Endocrine Neoplasia pathology, Pheochromocytoma metabolism, Pheochromocytoma pathology
- Abstract
Pheochromocytomas (PCCs) are tumors arising from neural crest-derived chromaffin cells. There are currently few animal models of PCC that recapitulate the key features of human tumors. Because such models may be useful for investigations of molecular pathomechanisms and development of novel therapeutic interventions, we characterized a spontaneous animal model (multiple endocrine neoplasia [MENX] rats) that develops endogenous PCCs with complete penetrance. Urine was longitudinally collected from wild-type (wt) and MENX-affected (mutant) rats and outputs of catecholamines and their O-methylated metabolites determined by mass spectrometry. Adrenal catecholamine contents, cellular ultrastructure, and expression of phenylethanolamine N-methyltransferase, which converts norepinephrine to epinephrine, were also determined in wt and mutant rats. Blood pressure was longitudinally measured and end-organ pathology assessed. Compared with wt rats, mutant animals showed age-dependent increases in urinary outputs of norepinephrine (P = .0079) and normetanephrine (P = .0014) that correlated in time with development of tumor nodules, increases in blood pressure, and development of hypertension-related end-organ pathology. Development of tumor nodules, which lacked expression of N-methyltransferase, occurred on a background of adrenal medullary morphological and biochemical changes occurring as early as 1 month of age and involving increased adrenal medullary concentrations of dense cored vesicles, tissue contents of both norepinephrine and epinephrine, and urinary outputs of metanephrine, the metabolite of epinephrine. Taken together, MENX-affected rats share several biochemical and pathophysiological features with PCC patients. This model thus provides a suitable platform to study the pathogenesis of PCC for preclinical translational studies aimed at the development of novel therapies for aggressive forms of human tumors.
- Published
- 2016
- Full Text
- View/download PDF
35. Animal models of multiple endocrine neoplasia.
- Author
-
Wiedemann T and Pellegata NS
- Subjects
- Animals, Disease Models, Animal, Female, Genetic Predisposition to Disease, Humans, Male, Mice, Multiple Endocrine Neoplasia genetics, Multiple Endocrine Neoplasia veterinary, Proto-Oncogene Mas, Rats, Cyclin-Dependent Kinase Inhibitor p27 genetics, Multiple Endocrine Neoplasia pathology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-ret genetics
- Abstract
Multiple endocrine neoplasia (MEN) syndromes are autosomal dominant diseases with high penetrance characterized by proliferative lesions (usually hyperplasia or adenoma) arising in at least two endocrine tissues. Four different MEN syndromes have been so far identified: MEN type 1 (MEN1), MEN2A (also referred to as MEN2), MEN2B (or MEN3) and MEN4, which have slightly varying tumor spectra and are caused by mutations in different genes. MEN1 associates with loss-of-function mutations in the MEN1 gene encoding the tumor suppressor menin. The MEN2A and MEN2B syndromes are due to activating mutations in the proto-oncogene RET (Rearranged in Transfection) and are characterized by different phenotypic features of the affected patients. MEN4 was the most recent addition to the family of the MEN syndromes. It was discovered less than 10 years ago thanks to studies of a rat strain that spontaneously develops multiple endocrine tumors (named MENX). These studies identified an inactivating mutation in the Cdkn1b gene, encoding the putative tumor suppressor p27, as the causative mutation of the rat syndrome. Subsequently, germline mutations in the human ortholog CDKN1B were also found in a subset of patients with a MEN-like phenotype and this led to the identification of MEN4. Small animal models have been instrumental in understanding important biochemical, physiological and pathological processes of cancer onset and spread in intact living organisms. Moreover, they have provided us with insight into gene function(s) and molecular mechanisms of disease progression. We here review the currently available animal models of MEN syndromes and their impact on the elucidation of the pathophysiology of these diseases, with a special focus on the rat MENX syndrome that we have been characterizing., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. Aggressive tumor growth and clinical evolution in a patient with X-linked acro-gigantism syndrome.
- Author
-
Naves LA, Daly AF, Dias LA, Yuan B, Zakir JC, Barra GB, Palmeira L, Villa C, Trivellin G, Júnior AJ, Neto FF, Liu P, Pellegata NS, Stratakis CA, Lupski JR, and Beckers A
- Subjects
- Adenoma pathology, Adenoma surgery, Child, Genetic Diseases, X-Linked pathology, Genetic Diseases, X-Linked surgery, Gigantism pathology, Gigantism surgery, Humans, Male, Pituitary Neoplasms pathology, Pituitary Neoplasms surgery, Treatment Outcome, Adenoma genetics, Genetic Diseases, X-Linked genetics, Gigantism genetics, Pituitary Neoplasms genetics
- Abstract
X-linked acro-gigantism (X-LAG) syndrome is a newly described disease caused by microduplications on chromosome Xq26.3 leading to copy number gain of GPR101. We describe the clinical progress of a sporadic male X-LAG syndrome patient with an Xq26.3 microduplication, highlighting the aggressive natural history of pituitary tumor growth in the absence of treatment. The patient first presented elsewhere aged 5 years 8 months with a history of excessive growth for >2 years. His height was 163 cm, his weight was 36 kg, and he had markedly elevated GH and IGF-1. MRI showed a non-invasive sellar mass measuring 32.5 × 23.9 × 29.1 mm. Treatment was declined and the family was lost to follow-up. At the age of 10 years and 7 months, he presented again with headaches, seizures, and visual disturbance. His height had increased to 197 cm. MRI showed an invasive mass measuring 56.2 × 58.1 × 45.0 mm, with compression of optic chiasma, bilateral cavernous sinus invasion, and hydrocephalus. His thyrotrope, corticotrope, and gonadotrope axes were deficient. Surgery, somatostatin analogs, and cabergoline did not control vertical growth and pegvisomant was added, although vertical growth continues (currently 207 cm at 11 years 7 months of age). X-LAG syndrome is a new genomic disorder in which early-onset pituitary tumorigenesis can lead to marked overgrowth and gigantism. This case illustrates the aggressive nature of tumor evolution and the challenging clinical management in X-LAG syndrome.
- Published
- 2016
- Full Text
- View/download PDF
37. Obesity in MENX Rats Is Accompanied by High Circulating Levels of Ghrelin and Improved Insulin Sensitivity.
- Author
-
Wiedemann T, Bielohuby M, Müller TD, Bidlingmaier M, and Pellegata NS
- Subjects
- Agouti-Related Protein metabolism, Animals, Appetite Regulation drug effects, Appetite Regulation physiology, Blood Glucose metabolism, Cyclin-Dependent Kinase Inhibitor p27 genetics, Hypothalamus metabolism, Multiple Endocrine Neoplasia complications, Multiple Endocrine Neoplasia genetics, Mutation, Neuropeptide Y metabolism, Obesity complications, RNA, Messenger metabolism, Rats, Receptors, Ghrelin antagonists & inhibitors, Receptors, Ghrelin metabolism, Receptors, Ghrelin therapeutic use, Ghrelin blood, Insulin Resistance physiology, Multiple Endocrine Neoplasia blood, Obesity blood
- Abstract
Ghrelin, the natural ligand of the growth hormone secretagogue receptor type 1a (GHS-R1a), is mainly secreted from the stomach and regulates food intake and energy homeostasis. p27 regulates cell cycle progression in many cell types. Here, we report that rats affected by the multiple endocrine neoplasia syndrome MENX, caused by a p27 mutation, develop pancreatic islet hyperplasia containing elevated numbers of ghrelin-producing ε-cells. The metabolic phenotype of MENX-affected rats featured high endogenous acylated and unacylated plasma ghrelin levels. Supporting increased ghrelin action, MENX rats show increased food intake, enhanced body fat mass, and elevated plasma levels of triglycerides and cholesterol. Ghrelin effect on food intake was confirmed by treating MENX rats with a GHS-R1a antagonist. At 7.5 months, MENX-affected rats show decreased mRNA levels of hypothalamic GHS-R1a, neuropeptide Y (NPY), and agouti-related protein (AgRP), suggesting that prolonged hyperghrelinemia may lead to decreased ghrelin efficacy. In line with ghrelin's proposed role in glucose metabolism, we find decreased glucose-stimulated insulin secretion in MENX rats, while insulin sensitivity is improved. In summary, we provide a novel nontransgenic rat model with high endogenous ghrelin plasma levels and, interestingly, improved glucose tolerance. This model might aid in identifying new therapeutic approaches for obesity and obesity-related diseases, including type 2 diabetes., (© 2016 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.)
- Published
- 2016
- Full Text
- View/download PDF
38. Oncogenic features of the bone morphogenic protein 7 (BMP7) in pheochromocytoma.
- Author
-
Leinhäuser I, Richter A, Lee M, Höfig I, Anastasov N, Fend F, Ercolino T, Mannelli M, Gimenez-Roqueplo AP, Robledo M, de Krijger R, Beuschlein F, Atkinson MJ, and Pellegata NS
- Subjects
- Adrenal Gland Neoplasms genetics, Animals, Apoptosis physiology, Case-Control Studies, Cell Proliferation physiology, Cohort Studies, Female, Humans, Male, PC12 Cells, Pheochromocytoma genetics, Phosphatidylinositol 3-Kinases genetics, Rats, Signal Transduction, Transfection, Adrenal Gland Neoplasms metabolism, Bone Morphogenetic Protein 7 metabolism, Pheochromocytoma metabolism, Phosphatidylinositol 3-Kinases metabolism
- Abstract
BMP7 is a growth factor playing pro- or anti-oncogenic roles in cancer in a cell type-dependent manner. We previously reported that the BMP7 gene is overexpressed in pheochromocytomas (PCCs) developing in MENX-affected rats and human patients. Here, analyzing a large cohort of PCC patients, we found that 72% of cases showed elevated levels of the BMP7 protein. To elucidate the role of BMP7 in PCC, we modulated its levels in PCC cell lines (overexpression in PC12, knockdown in MPC and MTT cells) and conducted functional assays. Active BMP signaling promoted cell proliferation, migration, and invasion, and sustained survival of MENX rat primary PCC cells. In PCC, BMP7 signals through the PI3K/AKT/mTOR pathway and causes integrin β1 up-regulation. Silencing integrin β1 in PC12 cells suppressed BMP7-mediated oncogenic features. Treatment of MTT cells with DMH1, a novel BMP antagonist, suppressed proliferation and migration. To verify the clinical applicability of our findings, we evaluated a dual PI3K/mTOR inhibitor (NVP-BEZ235) in MENX-affected rats in vivo. PCCs treated with NVP-BEZ235 had decreased proliferation and integrin β1 levels, and higher apoptosis. Altogether, BMP7 activates pro-oncogenic pathways in PCC. Downstream effectors of BMP7-mediated signaling may represent novel targets for treating progressive/inoperable PCC, still orphan of effective therapy.
- Published
- 2015
- Full Text
- View/download PDF
39. Analysis of mammalian gene function through broad-based phenotypic screens across a consortium of mouse clinics.
- Author
-
de Angelis MH, Nicholson G, Selloum M, White J, Morgan H, Ramirez-Solis R, Sorg T, Wells S, Fuchs H, Fray M, Adams DJ, Adams NC, Adler T, Aguilar-Pimentel A, Ali-Hadji D, Amann G, André P, Atkins S, Auburtin A, Ayadi A, Becker J, Becker L, Bedu E, Bekeredjian R, Birling MC, Blake A, Bottomley J, Bowl M, Brault V, Busch DH, Bussell JN, Calzada-Wack J, Cater H, Champy MF, Charles P, Chevalier C, Chiani F, Codner GF, Combe R, Cox R, Dalloneau E, Dierich A, Di Fenza A, Doe B, Duchon A, Eickelberg O, Esapa CT, El Fertak L, Feigel T, Emelyanova I, Estabel J, Favor J, Flenniken A, Gambadoro A, Garrett L, Gates H, Gerdin AK, Gkoutos G, Greenaway S, Glasl L, Goetz P, Da Cruz IG, Götz A, Graw J, Guimond A, Hans W, Hicks G, Hölter SM, Höfler H, Hancock JM, Hoehndorf R, Hough T, Houghton R, Hurt A, Ivandic B, Jacobs H, Jacquot S, Jones N, Karp NA, Katus HA, Kitchen S, Klein-Rodewald T, Klingenspor M, Klopstock T, Lalanne V, Leblanc S, Lengger C, le Marchand E, Ludwig T, Lux A, McKerlie C, Maier H, Mandel JL, Marschall S, Mark M, Melvin DG, Meziane H, Micklich K, Mittelhauser C, Monassier L, Moulaert D, Muller S, Naton B, Neff F, Nolan PM, Nutter LM, Ollert M, Pavlovic G, Pellegata NS, Peter E, Petit-Demoulière B, Pickard A, Podrini C, Potter P, Pouilly L, Puk O, Richardson D, Rousseau S, Quintanilla-Fend L, Quwailid MM, Racz I, Rathkolb B, Riet F, Rossant J, Roux M, Rozman J, Ryder E, Salisbury J, Santos L, Schäble KH, Schiller E, Schrewe A, Schulz H, Steinkamp R, Simon M, Stewart M, Stöger C, Stöger T, Sun M, Sunter D, Teboul L, Tilly I, Tocchini-Valentini GP, Tost M, Treise I, Vasseur L, Velot E, Vogt-Weisenhorn D, Wagner C, Walling A, Weber B, Wendling O, Westerberg H, Willershäuser M, Wolf E, Wolter A, Wood J, Wurst W, Yildirim AÖ, Zeh R, Zimmer A, Zimprich A, Holmes C, Steel KP, Herault Y, Gailus-Durner V, Mallon AM, and Brown SD
- Subjects
- Animals, Female, Heterozygote, Homozygote, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Molecular Sequence Annotation, Mutation, Phenotype, Genetic Association Studies
- Abstract
The function of the majority of genes in the mouse and human genomes remains unknown. The mouse embryonic stem cell knockout resource provides a basis for the characterization of relationships between genes and phenotypes. The EUMODIC consortium developed and validated robust methodologies for the broad-based phenotyping of knockouts through a pipeline comprising 20 disease-oriented platforms. We developed new statistical methods for pipeline design and data analysis aimed at detecting reproducible phenotypes with high power. We acquired phenotype data from 449 mutant alleles, representing 320 unique genes, of which half had no previous functional annotation. We captured data from over 27,000 mice, finding that 83% of the mutant lines are phenodeviant, with 65% demonstrating pleiotropy. Surprisingly, we found significant differences in phenotype annotation according to zygosity. New phenotypes were uncovered for many genes with previously unknown function, providing a powerful basis for hypothesis generation and further investigation in diverse systems.
- Published
- 2015
- Full Text
- View/download PDF
40. Acromegaly due to a Macroinvasive Plurihormonal Pituitary Adenoma and a Rectal Carcinoid Tumor.
- Author
-
Chin SO, Hwang JK, Rhee SY, Chon S, Oh S, Lee M, Pellegata NS, and Kim SW
- Abstract
A macroinvasive pituitary adenoma with plurihormonality usually causes acromegaly and hyperprolactinemia, and also accompanies with neurologic symptoms such as visual disturbances. However, its concurrent presentation with a rectal carcinoid tumor is rarely observed. This study reports the history, biochemical, colonoscopic and immunohistochemical results of a 48-year-old female with acromegaly and hyperprolactinemia. Despite the large size and invasive nature of the pituitary adenoma to adjacent anatomical structures, she did not complain of any neurologic symptoms such as visual disturbance or headache. Immunohistochemical staining of the surgical specimen from the pituitary adenoma revealed that the tumor cells were positive for growth hormone (GH), prolactin (PRL), and thyroid stimulating hormone (TSH). Staining for pituitary-specific transcription factor-1 (Pit-1) was shown to be strongly positive, which could have been possibly contributing to the plurihormonality of this adenoma. Colonoscopy found a rectal polyp that was identified to be a carcinoid tumor using immunohistochemical staining. A macroinvasive pituitary adenoma with concomitant rectal carcinoid tumor was secreting GH, PRL, and TSH, which were believed to be in association with over-expression of Pit-1. This is the first case report of double primary tumors comprising a plurihormonal pituitary macroadenoma and rectal carcinoid tumor.
- Published
- 2015
- Full Text
- View/download PDF
41. Targeting PI3K/mTOR Signaling Displays Potent Antitumor Efficacy against Nonfunctioning Pituitary Adenomas.
- Author
-
Lee M, Wiedemann T, Gross C, Leinhäuser I, Roncaroli F, Braren R, and Pellegata NS
- Subjects
- Animals, Defensins biosynthesis, Disease Models, Animal, Fluorescent Antibody Technique, Gene Expression Regulation drug effects, Humans, Rats, Rats, Mutant Strains, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, beta-Defensins biosynthesis, Antineoplastic Agents pharmacology, Imidazoles pharmacology, Phosphoinositide-3 Kinase Inhibitors, Pituitary Neoplasms pathology, Quinolines pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Purpose: Novel therapeutic approaches are needed to improve the postoperative management of residual nonfunctioning pituitary adenomas (NFPA), given their high relapse rate. Here, we evaluated the antitumor efficacy of the dual PI3K/mTOR inhibitor NVP-BEZ235 in the only available model of spontaneous NFPAs (MENX rats)., Experimental Design: Organotypic cultures of rat primary NFPAs were incubated with NVP-BEZ235 and assessed for cell viability, proliferation, apoptosis, and PI3K/mTOR inhibition. NVP-BEZ235, or placebo, was administered to MENX rats and tumor response was monitored noninvasively by diffusion weighted-magnetic resonance imaging (DW-MRI). Following treatment, tumor tissues were investigated for cell proliferation, apoptosis, and PI3K/mTOR inhibition. Genes mediating the cytotoxic activity of NVP-BEZ235 were identified by gene-expression profiling. Among them, Defb1, encoding beta-defensin 1, was further studied for its role in pituitary cells and in human pancreatic neuroendocrine tumor (NET) cells., Results: NVP-BEZ235 showed antiproliferative and pro-cell death activities against NFPAs both in vitro and in vivo, and the response to the drug correlated with inhibition of the PI3K pathway. DW-MRI identified early functional changes (decreased cellularity) in the adenomas before their size was affected and emerged as a useful modality to assess therapy response. The cytotoxic effect of PI3K/mTOR blockade in NFPA was mediated by several genes, including Defb1. NVP-BEZ235 treatment induced Defb1 expression in NFPAs in vitro and in vivo, and in pancreatic NET cells. High Defb1 levels sensitized NET cells to PI3K/mTOR inhibition., Conclusions: Our findings provide rationale for clinical investigation of PI3K/mTOR inhibition in NFPAs and identify novel effectors of PI3K-mediated neuroendocrine cell survival., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
42. Adrenomedullary progenitor cells: Isolation and characterization of a multi-potent progenitor cell population.
- Author
-
Vukicevic V, Rubin de Celis MF, Pellegata NS, Bornstein SR, Androutsellis-Theotokis A, and Ehrhart-Bornstein M
- Subjects
- Adrenal Medulla transplantation, Animals, Carcinogenesis pathology, Humans, Models, Biological, Stem Cell Transplantation, Adrenal Medulla cytology, Cell Separation methods, Multipotent Stem Cells cytology
- Abstract
The adrenal is a highly plastic organ with the ability to adjust to physiological needs by adapting hormone production but also by generating and regenerating both adrenocortical and adrenomedullary tissue. It is now apparent that many adult tissues maintain stem and progenitor cells that contribute to their maintenance and adaptation. Research from the last years has proven the existence of stem and progenitor cells also in the adult adrenal medulla throughout life. These cells maintain some neural crest properties and have the potential to differentiate to the endocrine and neural lineages. In this article, we discuss the evidence for the existence of adrenomedullary multi potent progenitor cells, their isolation and characterization, their differentiation potential as well as their clinical potential in transplantation therapies but also in pathophysiology., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
43. Semiquantitative 123I-Metaiodobenzylguanidine Scintigraphy to Distinguish Pheochromocytoma and Paraganglioma from Physiologic Adrenal Uptake and Its Correlation with Genotype-Dependent Expression of Catecholamine Transporters.
- Author
-
van Berkel A, Rao JU, Lenders JW, Pellegata NS, Kusters B, Piscaer I, Hermus AR, Plantinga TS, Langenhuijsen JF, Vriens D, Janssen MJ, Gotthardt M, and Timmers HJ
- Subjects
- Adolescent, Adrenal Gland Neoplasms diagnosis, Adrenal Gland Neoplasms genetics, Adult, Aged, Catecholamines metabolism, Cell Membrane diagnostic imaging, Child, Chromaffin Cells diagnostic imaging, Female, Genotype, Humans, Liver microbiology, Male, Middle Aged, Mutation, Neurofibromin 1 genetics, Norepinephrine Plasma Membrane Transport Proteins metabolism, Paraganglioma diagnosis, Paraganglioma genetics, Pheochromocytoma diagnosis, Pheochromocytoma genetics, Proto-Oncogene Proteins c-ret genetics, Retrospective Studies, Succinate Dehydrogenase genetics, Tomography, Emission-Computed, Single-Photon, Vesicular Monoamine Transport Proteins metabolism, Von Hippel-Lindau Tumor Suppressor Protein genetics, Young Adult, 3-Iodobenzylguanidine, Adrenal Gland Neoplasms diagnostic imaging, Adrenal Glands diagnostic imaging, Paraganglioma diagnostic imaging, Pheochromocytoma diagnostic imaging, Radionuclide Imaging methods
- Abstract
Unlabelled: (123)I-metaiodobenzylguanidine ((123)I-MIBG) scintigraphy plays an important role in the diagnostic evaluation of patients with pheochromocytoma and paraganglioma (PPGL). (123)I-MIBG targets cell membrane and vesicular catecholamine transporters of chromaffin cells and facilitates localization of the primary tumor and metastatic lesions. Its specificity for the diagnosis of adrenomedullary chromaffin cell tumors can be jeopardized by physiologic uptake by the normal adrenal medulla. The aim of this study was to distinguish between PPGLs and normal adrenal glands by evaluating semiquantitative (123)I-MIBG uptake and to examine genotype-specific differences in correlation with expression of catecholamine transporter systems., Methods: Sixty-two PPGLs collected from 57 patients with hereditary mutations in SDHA (n = 1), SDHB (n = 2), and SDHD (n = 4) (SDH is succinate dehydrogenase); von Hippel-Lindau (VHL; n = 2); RET (n = 12); neurofibromin 1 (NF1; n = 2); and MYC-associated factor X (MAX; n = 1), and with sporadic PPGLs (n = 33) were investigated. Preoperative planar and SPECT images were semiquantitatively analyzed using uptake measurements. Tumor-to-liver and normal adrenal-to-liver ratios were calculated and correlated with clinical characteristics including genotype, tumor size, and plasma metanephrines concentrations. The expression of norepinephrine transporter (NET) and vesicular monoamine transporter (VMAT-1) was evaluated immunohistochemically in paraffin-embedded tumor tissues., Results: Mean tumor-to-liver ratios of PPGL lesions were significantly higher than normal adrenal-to-liver ratios (P < 0.001). Cutoff values to distinguish between physiologic and pathologic adrenal uptake were established at 0.7 (100% sensitivity, 10.3% specificity) and 4.3 (100% specificity, 66.1% sensitivity). No statistically significant differences in (123)I-MIBG uptake were found across PPGLs of different genotypes. Mean NET expression in hereditary cluster 2 (RET, NF1, MAX) and apparently sporadic tumors was significantly higher than for hereditary cluster 1 (SDHx, VHL) PPGLs (P = 0.011 and 0.006, respectively). Mean VMAT-1 expression in hereditary cluster 1 PPGLs was significantly higher than for cluster 2 tumors (P = 0.010). (123)I-MIBG uptake significantly correlated with maximum tumor diameter (P = 0.002). (123)I-MIBG uptake, however, did not correlate with either NET or VMAT-1 expression., Conclusion: Liver-normalized semiquantitative (123)I-MIBG uptake may be helpful to distinguish between pheochromocytoma and physiologic adrenal uptake. Genotype-specific differences in the expression of NET and VMAT-1 do not translate into differences in (123)I-MIBG uptake., (© 2015 by the Society of Nuclear Medicine and Molecular Imaging, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
44. Early onset acromegaly associated with a novel deletion in CDKN1B 5'UTR region.
- Author
-
Sambugaro S, Di Ruvo M, Ambrosio MR, Pellegata NS, Bellio M, Guerra A, Buratto M, Foschini MP, Tagliati F, degli Uberti E, and Zatelli MC
- Subjects
- 5' Untranslated Regions, Adult, Age of Onset, Female, Gene Deletion, Humans, Acromegaly genetics, Cyclin-Dependent Kinase Inhibitor p27 genetics
- Abstract
Genetic alterations frequently are involved in the development of a pituitary adenoma in young age. We here characterize the functional role of a deletion in CDKN1B 5'-UTR region (c.-29_-26delAGAG) identified in an acromegalic patient that developed a growth hormone in pituitary adenoma during childhood. Our results show that the identified novel heterozygous deletion in the CDKN1B 5'-UTR region associates with a reduction in CDKN1B mRNA levels, a predicted altered secondary mRNA structure, and a reduced CDKN1B 5'-UTR transcriptional activity in vitro. The patient displayed loss of heterozygosity in the same CDKN1B 5'-UTR region at tissue level and the 5'UTR region containing the deleted sequence encompasses a GRE. These findings indicate that the identification of functional alterations of newly discovered genetic derangements need to be fully characterized and always correlated with the clinical manifestations.
- Published
- 2015
- Full Text
- View/download PDF
45. Functional characterization of a CDKN1B mutation in a Sardinian kindred with multiple endocrine neoplasia type 4 (MEN4).
- Author
-
Pardi E, Mariotti S, Pellegata NS, Benfini K, Borsari S, Saponaro F, Torregrossa L, Cappai A, Satta C, Mastinu M, Marcocci C, and Cetani F
- Abstract
Inactivating germline mutations of the CDKN1B gene, encoding for the nuclear cyclin-dependent kinase inhibitor p27kip1 protein, have been reported in patients with multiple endocrine neoplasia type 4 (MEN4), a MEN1-like phenotype without MEN1 mutations. The aim of this study was to in vitro characterize the germline CDKN1B mutation c.374_375delCT (S125X) we detected in a patient with MEN4. The proband was affected by multiglandular primary hyperparathyroidism and gastro-entero-pancreatic tumors. We carried out subcellular localization experiments transfecting into eukaryotic HeLa and GH3 cell lines plasmid vectors expressing the CDKN1B wild type (wt) or mutant cDNA. Western blot studies showed that fusion proteins were expressed at equal levels. The mutated protein was shorter compared to the wt protein and lacked the highly conserved C-terminal domain, which includes the bipartite nuclear localization signal at amino acids 152/153 and 166/168. In HeLa and GH3 cells wt p27 localized in the nucleus whereas the p27_S125X protein was retained in the cytoplasm predicting the loss of tumor suppressive function. The proband's tumoral parathyroid tissue did not show allelic loss, since wt and mutant alleles were both present by sequencing the somatic DNA. Immunohistochemistry showed a complete loss of nuclear p27 expression in the parathyroid adenoma removed by the patient at the second surgery. In conclusion, our study confirms the pathogenic role of the c.374_375delCT CDKN1B germline mutation in a patient with MEN4.
- Published
- 2015
- Full Text
- View/download PDF
46. SSTR3 is a putative target for the medical treatment of gonadotroph adenomas of the pituitary.
- Author
-
Lee M, Lupp A, Mendoza N, Martin N, Beschorner R, Honegger J, Schlegel J, Shively T, Pulz E, Schulz S, Roncaroli F, and Pellegata NS
- Subjects
- Adenoma metabolism, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Molecular Targeted Therapy, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local metabolism, Pituitary Neoplasms metabolism, Somatostatin therapeutic use, Young Adult, Adenoma drug therapy, Adenoma genetics, Pituitary Neoplasms drug therapy, Pituitary Neoplasms genetics, Receptors, Somatostatin metabolism, Somatostatin analogs & derivatives
- Abstract
Gonadotroph pituitary adenomas (GPAs) often present as invasive macroadenomas not amenable to complete surgical resection. Radiotherapy is the only post-operative option for patients with large invasive or recurrent lesions. No medical treatment is available for these patients. The somatostatin analogs (SSAs) octreotide and lanreotide that preferentially target somatostatin receptor type 2 (SSTR2) have little effect on GPAs. It is widely accepted that the expression of specific SSTR subtypes determines the response to SSAs. Given that previous studies on mRNA and protein expression of SSTRs in GPAs have generated conflicting results, we investigated the expression of SSTR2, SSTR3, and SSTR5 (the main targets of available SSAs) in a clinically and pathologically well-characterized cohort of 108 patients with GPAs. A total of 118 samples were examined by immunohistochemistry using validated and specific MABs. Matched primary and recurrent tissues were available for ten patients. The results obtained were validated in an independent cohort of 27 GPAs. We observed that SSTR3 was significantly more abundant than SSTR2 (P<0.0001) in GPAs, while full-length SSTR5 was only expressed in few tumors. Expression of SSTR3 was similar in primary and recurrent adenomas, was high in potentially aggressive lesions, and did not change significantly in adenomas that recurred after irradiation. In conclusion, low levels of expression of SSTR2 may account for the limited response of GPAs to octreotide and lanreotide. Given the potent anti-proliferative, pro-apoptotic, and anti-angiogenic activities of SSTR3, targeting this receptor with a multireceptor ligand SSA such as pasireotide may be indicated for potentially aggressive GPAs., (© 2015 Society for Endocrinology.)
- Published
- 2015
- Full Text
- View/download PDF
47. Ovarian hyperstimulation from ectopic hypersecretion of follicle stimulating hormone.
- Author
-
Miras AD, Mogford JT, Wright J, Mendoza NN, Xekouki P, Lakhani A, Pellegata NS, Stratakis CA, Roncaroli F, and Russell-Jones D
- Subjects
- Adenoma complications, Adenoma diagnosis, Adenoma surgery, Adult, Diagnosis, Differential, Female, Humans, Incidental Findings, Neuroendocrine Tumors complications, Neuroendocrine Tumors diagnosis, Neuroendocrine Tumors surgery, Pancreatic Neoplasms complications, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms surgery, Pituitary Neoplasms complications, Pituitary Neoplasms diagnosis, Pituitary Neoplasms surgery, Follicle Stimulating Hormone physiology, Ovarian Hyperstimulation Syndrome etiology
- Published
- 2015
- Full Text
- View/download PDF
48. Association between the p27 rs2066827 variant and tumor multiplicity in patients harboring MEN1 germline mutations.
- Author
-
Longuini VC, Lourenço DM Jr, Sekiya T, Meirelles O, Goncalves TD, Coutinho FL, Francisco G, Osaki LH, Chammas R, Alves VA, Siqueira SA, Schlesinger D, Naslavsky MS, Zatz M, Duarte YA, Lebrão ML, Gama P, Lee M, Molatore S, Pereira MA, Jallad RS, Bronstein MD, Cunha-Neto MB, Liberman B, Fragoso MC, Toledo SP, Pellegata NS, and Toledo RA
- Subjects
- Adolescent, Adult, Aged, Cohort Studies, Female, Humans, Male, Middle Aged, Multiple Endocrine Neoplasia Type 1 diagnosis, Young Adult, Cyclin-Dependent Kinase Inhibitor p27 genetics, Genetic Association Studies methods, Genetic Variation genetics, Germ-Line Mutation genetics, Multiple Endocrine Neoplasia Type 1 genetics, Proto-Oncogene Proteins genetics
- Abstract
Objective: To date, no evidence of robust genotype-phenotype correlation or disease modifiers for multiple endocrine neoplasia type 1 (MEN1) syndrome has been described, leaving the highly variable clinical presentation of patients unaccounted for., Design: As the CDKN1B (p27) gene causes MEN4 syndrome and it is transcriptionally regulated by the product of the MEN1 gene (menin), we sought to analyze whether p27 influences the phenotype of MEN1-mutated patients. The cohort consisted of 100 patients carrying germline MEN1 gene mutations and 855 population-matched control individuals., Methods: Genotyping of the coding p27 c.326T>G (V109G) variant was performed by sequencing and restriction site digestion, and the genotypes were associated with clinical parameters by calculating odds ratios (ORs) and their 95% CIs using logistic regression., Results: There were significant differences in p27 V109G allele frequencies between controls and MEN1-mutated patients (OR=2.55, P=0.019, CI=1.013-5.76). Among patients who are ≥30 years old carrying truncating MEN1 mutations, the T allele was strongly associated with susceptibility to tumors in multiple glands (three to four glands affected vs one to two glands affected; OR=18.33; P=0.002, CI=2.88-16.41). This finding remained significant after the Bonferroni's multiple testing correction, indicating a robust association. No correlations were observed with the development of MEN1-related tumors such as hyperparathyroidism, pituitary adenomas, and enteropancreatic and adrenocortical tumors., Conclusions: Our study suggests that the p27 tumor suppressor gene acts as a disease modifier for the MEN1 syndrome associated with MEN1 germline mutations. If confirmed in independent patient cohorts, this finding could facilitate the management of this clinically complex disease., (© 2014 European Society of Endocrinology.)
- Published
- 2014
- Full Text
- View/download PDF
49. p27 variant and corticotropinoma susceptibility: a genetic and in vitro study.
- Author
-
Sekiya T, Bronstein MD, Benfini K, Longuini VC, Jallad RS, Machado MC, Goncalves TD, Osaki LH, Higashi L, Viana J Jr, Kater C, Lee M, Molatore S, Francisco G, Chammas R, Naslavsky MS, Schlesinger D, Gama P, Duarte YA, Lebrão ML, Zatz M, Meirelles O, Liberman B, Fragoso MC, Toledo SP, Pellegata NS, and Toledo RA
- Subjects
- Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms metabolism, Adrenal Gland Neoplasms pathology, Adult, Aged, Animals, Apoptosis, Blotting, Western, Carcinoma, Neuroendocrine, Case-Control Studies, Cohort Studies, Female, Fluorescent Antibody Technique, Humans, In Vitro Techniques, Male, Mice, Middle Aged, Multiple Endocrine Neoplasia genetics, Multiple Endocrine Neoplasia metabolism, Multiple Endocrine Neoplasia pathology, Parathyroid Neoplasms metabolism, Parathyroid Neoplasms pathology, Pheochromocytoma metabolism, Pheochromocytoma pathology, Pituitary Neoplasms metabolism, Pituitary Neoplasms pathology, Rats, Thyroid Neoplasms metabolism, Thyroid Neoplasms pathology, Tumor Cells, Cultured, Tumor Stem Cell Assay, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p27 genetics, Mutation genetics, Parathyroid Neoplasms genetics, Pheochromocytoma genetics, Pituitary Neoplasms genetics, Thyroid Neoplasms genetics
- Abstract
Germline mutations in p27(kip1) are associated with increased susceptibility to multiple endocrine neoplasias (MEN) both in rats and humans; however, the potential role of common polymorphisms of this gene in endocrine tumor susceptibility and tumorigenesis remains mostly unrecognized. To assess the risk associated with polymorphism rs2066827 (p27-V109G), we genotyped a large cohort of Brazilian patients with sporadic endocrine tumors (pituitary adenomas, n=252; pheochromocytomas, n=125; medullary thyroid carcinoma, n=51; and parathyroid adenomas, n=19) and 885 population-matched healthy controls and determined the odds ratios and 95% CIs. Significant associations were found for the group of patients with pituitary adenomas (P=0.01), particularly for those with ACTH-secreting pituitary adenomas (P=0.005). In contrast, no association was found with GH-secreting pituitary tumors alone or with the sporadic counterpart of MEN2-component neoplasias. Our in vitro analyses revealed increased colony formation and cell growth rate for an AtT20 corticotropin mouse cell line overexpressing the p27-V109G variant compared with cells transfected with the WT p27. However, the genotypic effects in genetic and in vitro approaches were divergent. In accordance with our genetic data showing specificity for ACTH-secreting pituitary tissues, the overexpression of p27-V109G in a GH3 somatotropin rat cell line resulted in no difference compared with the WT. Pituitary tumors are one of the major clinical components of syndromes associated with the p27 pathogenic mutations MENX and MEN4. Our genetic and in vitro data indicate that the common polymorphism rs2066827 may play a role in corticotropinoma susceptibility and tumorigenesis through a molecular mechanism not fully understood thus far.
- Published
- 2014
- Full Text
- View/download PDF
50. Preclinical evaluation of 18F-LMI1195 for in vivo imaging of pheochromocytoma in the MENX tumor model.
- Author
-
Gaertner FC, Wiedemann T, Yousefi BH, Lee M, Repokis I, Higuchi T, Nekolla SG, Yu M, Robinson S, Schwaiger M, and Pellegata NS
- Subjects
- Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms pathology, Animals, Autoradiography, Disease Models, Animal, Feasibility Studies, Gene Expression Regulation, Neoplastic, Mutation, Norepinephrine Plasma Membrane Transport Proteins genetics, Organ Size, Pheochromocytoma genetics, Pheochromocytoma pathology, Rats, Tumor Burden, Adrenal Gland Neoplasms diagnostic imaging, Fluorine Radioisotopes, Fluorobenzenes pharmacokinetics, Guanidines pharmacokinetics, Multiple Endocrine Neoplasia, Pheochromocytoma diagnostic imaging, Positron-Emission Tomography
- Abstract
Unlabelled: We evaluated (18)F-LMI1195 (1-(3-bromo-4-(3-(18)F-fluoro-propoxy)benzyl)guanidine), a metaiodobenzylguanidine (MIBG) analog, for the detection of pheochromocytoma in a preclinical in vivo model of endogenous neuroendocrine tumors (multiple endocrine neoplasia [MENX])., Methods: Adrenal uptake kinetics of (18)F-LMI1195 were evaluated in healthy Wistar rats (n = 6) by dynamic PET imaging. Distribution of (18)F-LMI1195 was evaluated in tumor-bearing MENX mut/mut rats (n = 10) and control MENX wild-type rats (n = 4) by biodistribution studies and PET imaging. Biodistribution of (18)F-LMI1195 was compared with (123)I-MIBG in MENX mut/mut rats (n = 6) and correlated with histological tumor volume and norepinephrine transporter (NET) expression. Uptake specificity was evaluated by in vivo inhibition of the NET by desipramine (n = 6). Intraadrenal distribution of (18)F-LMI1195 was evaluated by autoradiography., Results: (18)F-LMI1195 showed rapid tracer accumulation in adrenal glands 1 min after tracer injection. Adrenal glands of MENX mut/mut animals showed significantly higher standardized uptake value than MENX wild-type controls (maximum SUV, 10.3 ± 2.3 vs. 6.1 ± 0.9, P < 0.01). Adrenal uptake in MENX mut/mut rats could be inhibited by desipramine, shown by biodistribution studies (0.06 ± 0.01 vs. 0.16 ± 0.05 percentage injected dose, P < 0.01), PET imaging (maximum SUV, 3.8 ± 0.8 vs. 10.3 ± 2.3, P < 0.01), and autoradiography. Adrenal uptake of (18)F-LMI1195 correlated with (123)I-MIBG uptake (r = 0.91), histological tumor volume (r = 0.68), and NET expression (r = 0.50). (18)F-LMI1195 showed an overall favorable distribution for tumor imaging., Conclusion: (18)F-LMI1195 shows high and specific accumulation in pheochromocytomas. Its favorable biodistribution makes it a promising PET tracer for tumor imaging. Further studies are warranted to evaluate its clinical value in oncologic indications.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.