Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Endocrinol Invest ; 47(3): 633-643, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37736856

ABSTRACT

PURPOSE: Indeterminate cytology still puzzles clinicians, due to its wide range of oncological risks. According to the Italian SIAPEC-IAP classification, TIR3B cytology holds up to 30% of thyroid cancer, which justifies the surgical indication, even if more than half of cases do not result in a positive histology. The study aim is to identify potential clinical, ultrasound or cytological features able to improve the surgical indication. METHODS: Retrospective analysis. A consecutive series of TIR3B nodules referred to the Endocrine Unit of Careggi Hospital from 1st May 2014 to 31st December 2021 was considered for the exploratory analysis (Phase 1). Thereafter, a smaller confirmatory sample of consecutive TIR3B diagnosed and referred to surgery from 1st January 2022 to 31st June 2022 was considered to verify the algorithm (Phase 2). The main clinical, ultrasound and cytological features have been collected. A comprehensive stepwise logistic regression was applied to build a prediction algorithm. The histological results represented the final outcome. RESULTS: Of 599 TIR3B nodules referred to surgery, 451 cases were included in the exploratory analysis. A final score > 14.5 corresponded to an OR = 4.98 (95% CI 3.24-7.65, p < 0.0001) and showed a PPV and NPV of 57% and 79%, respectively. The Phase 2 analysis on a confirmatory sample of 58 TIR3B cytology confirmed that a threshold of 14.5 points has a comparable PPV and NPV of 53% and 80%, respectively. CONCLUSIONS: A predictive algorithm which considers the main clinical, US and cytological features can significantly improve the oncological stratification of TIR3B cytology.


Subject(s)
Algorithms , Thyroid Neoplasms , Humans , Retrospective Studies , Hospitals , Medical Oncology
2.
J Endocrinol Invest ; 46(5): 967-983, 2023 May.
Article in English | MEDLINE | ID: mdl-36436190

ABSTRACT

PURPOSE: Hyponatremia is the most frequent electrolytic disorder in clinical practice. In addition to neurological symptoms, hyponatremia, even when mild/moderate and chronic, has been related to other manifestations, such as bone demineralization and increased risk of fractures. To better elucidate tissue alterations associated with reduced serum sodium concentration [Na+], we developed an in vivo model of hyponatremia secondary to the Syndrome of Inappropriate Antidiuresis. METHODS AND RESULTS: Hyponatremia was induced in Foxn1nu/nu mice by subcutaneous infusion of the vasopressin analog 1-deamino [8-D-arginine] vasopressin (dDAVP) for 14 days via osmotic mini-pumps. Mice in the control group were infused with isotonic saline solution. Serum [Na+] progressively decreased, with a nadir of 123.4 ± 2.3 mEq/L (mean ± SD, dDAVP 0.3 ng/h) and 111.6 ± 4.7 mEq/L (mean ± SD, dDAVP 0.5 ng/h). Evident signs of liver steatofibrosis were observed at histology in hyponatremic mice. Accordingly, the expression of proteins involved in lipid metabolism (SREBP-1, PPARα and PPARγ) and in myofibroblast formation (αSMA and CTGF) significantly increased. Furthermore, heme oxygenase 1 expression was up-regulated in Kupffer and hepatic stellate cells in the liver of hyponatremic mice. Testis alterations were also observed. In particular, the thickness of the seminiferous epithelium appeared reduced. The expression levels of PCNA and PTMA, which are involved in DNA replication and germ cells maturation, were markedly reduced in the testis of hyponatremic mice. CONCLUSION: Overall, these findings shed new light on the possible consequences of chronic hyponatremia and prompt a more thorough evaluation of hyponatremic patients.


Subject(s)
Hyponatremia , Inappropriate ADH Syndrome , Male , Mice , Animals , Hyponatremia/complications , Hyponatremia/diagnosis , Deamino Arginine Vasopressin , Inappropriate ADH Syndrome/complications , Inappropriate ADH Syndrome/diagnosis , Vasopressins , Sodium , Liver , Spermatogenesis
3.
J Endocrinol Invest ; 46(3): 577-586, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36284058

ABSTRACT

PURPOSE: Hyponatremia occurs in about 30% of patients with pneumonia, including those with SARS-CoV-2 (COVID-19) infection. Hyponatremia predicts a worse outcome in several pathologic conditions and in COVID-19 has been associated with a higher risk of non-invasive ventilation, ICU transfer and death. The main objective of this study was to determine whether early hyponatremia is also a predictor of long-term sequelae at follow-up. METHODS: In this observational study, we collected 6-month follow-up data from 189 laboratory-confirmed COVID-19 patients previously admitted to a University Hospital. About 25% of the patients (n = 47) had hyponatremia at the time of hospital admission. RESULTS: Serum [Na+] was significantly increased in the whole group of 189 patients at 6 months, compared to the value at hospital admission (141.4 ± 2.2 vs 137 ± 3.5 mEq/L, p < 0.001). In addition, IL-6 levels decreased and the PaO2/FiO2 increased. Accordingly, pulmonary involvement, evaluated at the chest X-ray by the RALE score, decreased. However, in patients with hyponatremia at hospital admission, higher levels of LDH, fibrinogen, troponin T and NT-ProBNP were detected at follow-up, compared to patients with normonatremia at admission. In addition, hyponatremia at admission was associated with worse echocardiography parameters related to right ventricular function, together with a higher RALE score. CONCLUSION: These results suggest that early hyponatremia in COVID-19 patients is associated with the presence of laboratory and imaging parameters indicating a greater pulmonary and right-sided heart involvement at follow-up.


Subject(s)
COVID-19 , Hyponatremia , Humans , COVID-19/complications , SARS-CoV-2 , Hyponatremia/complications , Follow-Up Studies , Respiratory Sounds , Hospitals , Retrospective Studies
4.
J Endocrinol Invest ; 45(9): 1693-1708, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35604542

ABSTRACT

PURPOSE: Hyponatremia, the most frequent electrolyte alteration in clinical practice, has been associated with a worse prognosis in cancer patients. On the other hand, a better outcome has been related to serum sodium normalization. In vitro studies have shown that low extracellular sodium promotes cancer cells proliferation and invasiveness. Tolvaptan, a selective vasopressin receptor type 2 (V2) antagonist, has been effectively used in the last decade for the treatment of hyponatremia secondary to the Syndrome of Inappropriate Antidiuresis. A few in vitro data suggested a direct role of tolvaptan in counteracting cancer progression, so far. The aim of this study was to evaluate the effect and the mechanism of action of tolvaptan in cell lines from different tumours [i.e. colon cancer (HCT-8), hepatocarcinoma (HepG2), neuroblastoma (SK-N-AS)]. METHODS AND RESULTS: First, we showed that these cell lines express the V2 receptor. Tolvaptan significantly reduced cell proliferation with an IC50 in the micromolar range. Accordingly, reduced levels of cAMP, of the catalytic α subunit of PKA, and a reduced pAKT/AKT ratio were observed. Tolvaptan effectively inhibited cell cycle progression, whereas it induced apoptotis. Furthermore, it reduced cell invasiveness. In particular, anchorage-independent growth and the activity of collagenases type IV were blunted in the three cell lines. Accordingly, tolvaptan counteracted the RhoA/ROCK1-2 pathway, which has a pivotal role in regulating cell movement. CONCLUSIONS: Overall, these findings indicate that tolvaptan effectively inhibits tumour progression in vitro. Further studies should clarify whether the V2 receptor might be considered a possible target in anti-cancer strategies in the future.


Subject(s)
Hyponatremia , Neoplasms , Antidiuretic Hormone Receptor Antagonists/pharmacology , Antidiuretic Hormone Receptor Antagonists/therapeutic use , Benzazepines/adverse effects , Cell Proliferation , Humans , Neoplasms/complications , Sodium , Tolvaptan/pharmacology , Tolvaptan/therapeutic use , rho-Associated Kinases/therapeutic use
5.
J Endocrinol Invest ; 43(9): 1259-1269, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32157664

ABSTRACT

PURPOSE: Nowadays, no human neuroendocrine cell models derived from the neural crest are available. In this study, we present non-transformed long-term primary Neural Crest Cells (NCCs) isolated from the trunk region of the neural crest at VIII-XII gestational weeks of human foetuses obtained from voluntary legal abortion. METHODS AND RESULTS: In NCC, quantitative real-time RT PCR demonstrated the expression of neural crest specifier genes, such as Snail1, Snail2/SLUG, Sox10, FoxD3, c-Myc, and p75NTR. Moreover, these cell populations expressed stemness markers (such as Nanog and nestin), as well as markers of motility and invasion (TAGLN, MMP9, CXCR4, and CXCR7), and of neuronal/glial differentiation (MAP2, GFAP, SYP, and TAU). Functional analysis demonstrated that these cells not only possessed high migration properties, but most importantly, they expressed markers of sympatho-adrenal lineage, such as ASCL1 and tyrosine hydroxylase (TH). Moreover, the expression of TH increased after the induction with two different protocols of differentiation towards neuronal and sympatho-adrenal phenotypes. Finally, exposure to conditioned culture media from NCC induced a mature phenotype in a neuronal cell model (namely SH-SY5Y), suggesting that NCC may also act like Schwann precursors. CONCLUSION: This unique human cell model provides a solid tool for future studies addressing the bases of human neural crest-derived neuroendocrine tumours.


Subject(s)
Cell Separation , Fetus/cytology , Neural Crest/cytology , Neuroendocrine Cells/cytology , Cell Differentiation , Cell Line , Cell Movement , Cell Separation/methods , Female , Humans , Neural Crest/embryology , Neural Crest/physiology , Neuroendocrine Cells/physiology , Phenotype , Pregnancy , Primary Cell Culture
6.
J Endocrinol Invest ; 39(2): 177-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26169632

ABSTRACT

BACKGROUND: Hyponatremia is associated with negative clinical outcomes even when chronic and mild. It is also known that hyponatremia treatment should be appropriately performed, to avoid dramatic consequences possibly leading to death. We have previously demonstrated that chronically low extracellular [Na(+)], independently of reduced osmolality, is associated with signs of neuronal cell distress, possibly involving oxidative stress. AIM: The aim of the present study was to assess whether the return to normal extracellular [Na(+)] is able to revert neuronal cell damage. METHODS: After exposing SH-SY5Y and SK-N-AS cells to low [Na(+)] and returning to normal [Na(+)], we analyzed cell viability by MTS assay, ROS accumulation by FASCan and expression of anti-apoptotic genes. RESULTS: We found that the viability of cells was restored upon return to normal [Na(+)]. However, when more subtle signs of cell distress were assessed, such as the expression level of the anti-apoptotic genes Bcl-2 and DHCR24 or of the heme oxygenase 1 gene, a complete return to basal values was not observed, in particular in SK-N-AS, even when [Na(+)] was gradually increased. We also demonstrated that the amount of ROS significantly increased in low [Na(+)], thus confirming that oxidative stress appears to contribute to the effects of low [Na(+)] on cell homeostasis. CONCLUSIONS: Overall, this study provided the first demonstration that the correction of chronically low extracellular [Na(+)] may not be able to revert all the cell alterations associated with reduced [Na(+)]. These results suggest that prompt hyponatremia treatment might prevent possible residual abnormalities.


Subject(s)
Gene Expression Regulation , Nerve Tissue Proteins/metabolism , Neurons/physiology , Osmoregulation , Oxidative Stress , Reactive Oxygen Species/metabolism , Stromal Cells/physiology , Biomarkers/metabolism , Cell Line , Cell Line, Tumor , Cell Survival , Extracellular Fluid/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hyponatremia/metabolism , Hyponatremia/therapy , Kinetics , Lipid Peroxidation , Nerve Tissue Proteins/genetics , Osmotic Pressure , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
7.
Endocrine ; 52(1): 73-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26093848

ABSTRACT

Hyponatremia represents an independent risk factor for osteoporosis and fractures, affecting both bone density and quality. A direct stimulation of bone resorption in the presence of reduced extracellular sodium concentrations ([Na(+)]) has been shown, but the effects of low [Na(+)] on osteoblasts have not been elucidated. We investigated the effects of a chronic reduction of extracellular [Na(+)], independently of osmotic stress, on human mesenchymal stromal cells (hMSC) from bone marrow, the common progenitor for osteoblasts and adipocytes. hMSC adhesion and viability were significantly inhibited by reduced [Na(+)], but their surface antigen profile and immuno-modulatory properties were not altered. In low [Na(+)], hMSC were able to commit toward both the osteogenic and the adipogenic phenotypes, as demonstrated by differentiation markers analysis. However, the dose-dependent increase in the number of adipocytes as a function of reduced [Na(+)] suggested a preferential commitment toward the adipogenic phenotype at the expense of osteogenesis. The amplified inhibitory effect on the expression of osteoblastic markers exerted by adipocytes-derived conditioned media in low [Na(+)] further supported this observation. The analysis of cytoskeleton showed that low [Na(+)] were associated with disruption of tubulin organization in hMSC-derived osteoblasts, thus suggesting a negative effect on bone quality. Finally, hMSC-derived osteoblasts increased their expression of factors stimulating osteoclast recruitment and activity. These findings confirm that hyponatremia should be carefully taken into account because of its negative effects on bone, in addition to the known neurological effects, and indicate for the first time that impaired osteogenesis may be involved.


Subject(s)
Adipogenesis , Bone Resorption/etiology , Bone Resorption/metabolism , Hyponatremia/complications , Hyponatremia/metabolism , Mesenchymal Stem Cells/metabolism , Sodium/deficiency , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Survival , Cytoskeleton/metabolism , Humans , Lymphocyte Culture Test, Mixed , Osmotic Pressure , Osteogenesis , Phenotype , Tubulin/metabolism
8.
J Clin Endocrinol Metab ; 98(8): 3359-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23788690

ABSTRACT

CONTEXT: Cell-free nucleic acids circulating in plasma are considered a promising noninvasive tool for cancer monitoring. BRAF(V600E) mutation in cell-free DNA (cfDNA) could represent an appropriate marker for papillary thyroid carcinoma (PTC). OBJECTIVE: Our aim is to investigate the role of BRAF(V600E)-mutated allele in cfDNA as a marker for the diagnosis and follow-up of PTC. STUDY DESIGN: BRAF(V600E) allele was detected and quantified by an allele-specific real-time quantitative PCR assay in plasma from 103 patients affected by nodular goiter. As control populations, we enrolled 49 healthy subjects and 16 patients with non-nodular thyroid diseases. RESULTS: The percentage of circulating BRAF(V600E) was significantly different between patients and controls and throughout different cytological categories of ultrasound-assisted fine-needle aspiration. Patients with a histopathological diagnosis of PTC showed a higher percentage of circulating BRAF(V600E) (P = .035) compared to those with benign histology. In 19 patients, a second blood draw, taken 3-6 months after surgery, showed a lower percentage of BRAF(V600E) in cfDNA than the presurgical sample (P < .001). The diagnostic performance of circulating BRAF(V600E) was assessed by receiver operating characteristic curve analysis resulting in an area under the curve of 0.797. A cutoff value was chosen corresponding to maximum specificity (65%) and sensitivity (80%). On this basis, we evaluated the predictive value of BRAF(V600E) in Thy 3 patients with a resulting positive predictive value of 33% and a negative predictive value of 80%. CONCLUSIONS: The results of the present study provide encouraging data supporting the possibility to take advantage of circulating BRAF(V600E) in the management of PTC.


Subject(s)
Carcinoma/diagnosis , Mutation , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma/blood , Carcinoma/genetics , Carcinoma/pathology , Carcinoma, Papillary , DNA/analysis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Proto-Oncogene Proteins B-raf/blood , ROC Curve , Thyroid Cancer, Papillary , Thyroid Neoplasms/blood , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
9.
J Neuroendocrinol ; 24(10): 1304-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22621285

ABSTRACT

Insulin-like growth factor-1 (IGF-1) and oestrogens interact with each other as neuroprotective factors. We have previously demonstrated that 17ß-oestradiol protects against ß-amyloid and oxidative stress toxicity and increases the amount of cell cholesterol in human foetal neuroblasts (FNC). The present study aimed: (i) to assess the protective effects of IGF-1 in FNC cells; (ii) to investigate the relationship between IGF-1 and 17ß-oestradiol; and (iii) to determine whether cholesterol was a major mediator of the effects of IGF-1, similarly to 17ß-oestradiol. We found that IGF-1 effectively exerts neuroprotective effects in FNC cells. We also demonstrated that the IGF-1 receptor (IGF-1R) pathway is needed to maintain oestrogen-mediated neuroprotection. Finally, we found that, opposite to 17ß-oestradiol, IGF-1 did not cause a significant increase in cell cholesterol. These findings indicate that a cross-talk between IGF-1 and 17ß-oestradiol occurs in FNC cells. In particular, the activation of the IGF-1R cascade appears to be fundamental to warrant 17ß-oestradiol-mediated neuroprotection, even though cell cholesterol does not play a major role as an effector of this pathway.


Subject(s)
Estradiol/pharmacology , Insulin-Like Growth Factor I/pharmacology , Neural Stem Cells/drug effects , Neuroprotective Agents , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholesterol/metabolism , Humans , Neural Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Receptor Cross-Talk/drug effects
10.
J Endocrinol Invest ; 33(10): 730-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20386089

ABSTRACT

BACKGROUND: Chronic inflammation is now considered a determinant of benign prostatic hyperplasia (BPH), promoting, together with the hormonal milieu, prostate overgrowth and lower urinary tract symptoms (LUTS). Prostatic urethra actively participates in determining progression of LUTS associated with BPH. AIM: To investigate the expression of the vitamin D receptor (VDR) and the ability of the VDR agonist elocalcitol to reduce inflammatory responses in human prostatic urethra (hPU) cells. MATERIALS AND METHODS: Human prostatic urethra, prostate and bladder neck were obtained from patients affected by BPH. Immunohistochemical studies for VDR expression were performed in tissue samples, from which primary cell cultures were also derived. In hPU cells, proliferation and chemiotaxis were studied, along with Rho kinase (ROCK) activity (MYPT-1 phosphorylation) by western blot. Quantitative RT-PCR was performed for VDR, cyclooxygenase (COX-2), and interleukin (IL)-8 expression. RESULTS: Urethra displays higher VDR expression compared to prostate and bladder neck tissues. The VDR agonist elocalcitol partially reverts COX-2 and IL-8 mRNA upregulation induced by a pro-inflammatory cytokine mixture (IL-17, interferon-γ, tumor necrosis factor-α) and inhibits cell migration in urethral cells. Elocalcitol prevents activation of ROCK, as previously demonstrated in bladder and prostate cell cultures. CONCLUSIONS: Our results suggest that prostatic urethra is, within the lower urinary tract, a novel target for VDR agonists, as shown by the capacity of elocalcitol to inhibit ROCK activity and to limit inflammatory responses in human primary urethra cells.


Subject(s)
Calcitriol/analogs & derivatives , Prostate/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/genetics , Urethra/metabolism , Aged , Calcitriol/pharmacology , Cell Culture Techniques , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Drug Evaluation, Preclinical , Gene Expression , Humans , Ligands , Male , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Prostate/drug effects , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/surgery , Receptors, Calcitriol/metabolism , Urethra/drug effects , Urethra/pathology , Urinary Bladder/metabolism , Urinary Bladder/pathology
11.
Int J Androl ; 33(3): 475-88, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-19508330

ABSTRACT

Benign prostatic hyperplasia (BPH) is a common disorder affecting 50-80% of the aged male population. Androgens and age have been traditionally considered the main determinants of prostate enlargement, but in the last years a potentially important role of chronic inflammation in BPH pathogenesis has emerged. Bacterial and non-infectious chronic prostatitis could represent inciting factors leading to tissue hyperproliferation, possibly via the recently demonstrated antigen-presenting capacity of prostatic stromal cells, enabling them to induce and sustain intraglandular immune responses. The prostate growth-promoting chemokine IL-8 could represent a direct link between chronic prostate inflammation and autocrine/paracrine stromal cell proliferation, in agreement with its marked secretion induced in BPH stromal cells by a combination of Th1 and Th17 cell-derived inflammatory cytokines. BPH stromal cells express the vitamin D receptor (VDR), which is up-regulated by exposure to inflammatory stimuli. The non-hypercalcaemic VDR agonist elocalcitol, shown to arrest BPH development by decreasing the intra-prostatic androgen signalling without directly interfering with systemic androgen action, exerts immunoregulatory and anti-inflammatory properties in different prostatic pathology characterized by growth and inflammation. The mechanism of action of VDR agonists supports an important role of chronic inflammation in BPH pathogenesis and strengthens the concept of these agents as a therapeutic option for pharmacological treatment of BPH.


Subject(s)
Inflammation/immunology , Prostatic Hyperplasia , Androgens/physiology , Androgens/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Calcitriol/analogs & derivatives , Chemokines/therapeutic use , Chronic Disease , Cytokines/immunology , Cytokines/therapeutic use , Humans , Inflammation/drug therapy , Interleukin-8/therapeutic use , Male , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Prostatic Hyperplasia/etiology , Prostatic Hyperplasia/pathology , Prostatitis/complications , Prostatitis/drug therapy , Prostatitis/pathology , Receptors, Calcitriol/agonists , Receptors, Calcitriol/metabolism , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...