29 results on '"Elena Lastraioli"'
Search Results
2. Editorial: Pharmacogenetic landscape in human solid cancers
- Author
-
Elena Lastraioli, Francesca Colombo, and Elisa Frullanti
- Subjects
pharmacogenetics ,solid cancer ,human tumours ,pharmacogenomics ,cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2024
- Full Text
- View/download PDF
3. Ion channels in lung cancer: biological and clinical relevance
- Author
-
Chiara Capitani, Ginevra Chioccioli Altadonna, Michele Santillo, and Elena Lastraioli
- Subjects
lung cancer ,SCLC ,NSCLC ,potassium channels ,sodium channels ,calcium channels ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Despite improvements in treatment, lung cancer is still a major health problem worldwide. Among lung cancer subtypes, the most frequent is represented by adenocarcinoma (belonging to the Non-Small Cell Lung Cancer class) although the most challenging and harder to treat is represented by Small Cell Lung Cancer, that occurs at lower frequency but has the worst prognosis. For these reasons, the standard of care for these patients is represented by a combination of surgery, radiation therapy and chemotherapy. In this view, searching for novel biomarkers that might help both in diagnosis and therapy is mandatory. In the last 30 years it was demonstrated that different families of ion channels are overexpressed in both lung cancer cell lines and primary tumours. The altered ion channel profile may be advantageous for diagnostic and therapeutic purposes since most of them are localised on the plasma membrane thus their detection is quite easy, as well as their block with specific drugs and antibodies. This review focuses on ion channels (Potassium, Sodium, Calcium, Chloride, Anion and Nicotinic Acetylcholine receptors) in lung cancer (both Non-Small Cell Lung Cancer and Small Cell Lung Cancer) and recapitulate the up-to-date knowledge about their role and clinical relevance for a potential use in the clinical setting, for lung cancer diagnosis and therapy.
- Published
- 2023
- Full Text
- View/download PDF
4. Corrigendum to 'Transcriptomic data of Bevacizumab-adapted colorectal adenocarcinoma cells HCT-116' [Data In Brief, Volume 48, (Available online 17 March 2023) 1-9/Article 109069]
- Author
-
Cesare Sala, Tiziano Lottini, Elena Lastraioli, Federico Alessandro Ruffinatti, Luca Visentin, and Annarosa Arcangeli
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Published
- 2023
- Full Text
- View/download PDF
5. hERG1 Potassium Channel Expression in Colorectal Adenomas: Comparison with Other Preneoplastic Lesions of the Gastrointestinal Tract
- Author
-
Elena Lastraioli, Jessica Iorio, Federica Petrelli, Anna Tomezzoli, Serena Battista, Maria Raffaella Ambrosio, Mariella Chiudinelli, Federica De Salvatore, Luca Messerini, Vincenzo Villanacci, Luca Saragoni, and Annarosa Arcangeli
- Subjects
hERG1 potassium channels ,colorectal adenomas ,gastric dysplasias ,Barrett’ s esophagus ,Biology (General) ,QH301-705.5 - Abstract
Preneoplastic lesions represent a useful target for early diagnosis and follow-up of gastrointestinal malignancies. hERG1 channel expression was tested by immunohistochemistry (IHC) in a cohort of colorectal adenoma samples belonging to Italian subjects. Overall, hERG1 was expressed in 56.5% of cases with both high staining intensity and a high percentage of positive cells. Moreover, hERG1 was expressed in a higher percentage of dysplastic adenomas with respect to hyperplastic lesions, and the proportion of positive samples further increased in patients with high-grade dysplasia. Comparing hERG1 expression in other preneoplastic lesions of the GI tract (gastric dysplasia and Barrett’s esophagus), it emerged that in all the conditions, hERG1 was expressed with a diffused pattern, throughout the cell, with variable staining intensity within the samples. The highest expression was detected in gastric dysplasia samples and the lowest in Barrett’s esophagus at similar levels observed in colorectal adenomas. Our results show that hERG1 is aberrantly expressed in human preneoplastic lesions of the gastrointestinal tract and has a different pattern of expression and role in the different sites. Overall, the detection of hERG1 expression in preneoplastic lesions could represent a novel diagnostic or prognostic marker of progression in the gastrointestinal tract.
- Published
- 2022
- Full Text
- View/download PDF
6. Transgenic mice overexpressing the LH receptor in the female reproductive system spontaneously develop endometrial tumour masses
- Author
-
Tiziano Lottini, Jessica Iorio, Elena Lastraioli, Laura Carraresi, Claudia Duranti, Cesare Sala, Miriam Armenio, Ivo Noci, Serena Pillozzi, and Annarosa Arcangeli
- Subjects
Medicine ,Science - Abstract
Abstract The receptor for the luteinizing hormone (LH-R) is aberrantly over expressed in cancers of the reproductive system. To uncover whether LH-R over expression has a causative role in cancer, we generated a transgenic (TG) mouse which overexpresses the human LH-R (hLH-R) in the female reproductive tract, under the control of the oviduct-specific glycoprotein (OGP) mouse promoter (mogp-1). The transgene was highly expressed in the uterus, ovary and liver, but only in the uterus morphological and molecular alterations (increased proliferation and trans-differentiation in the endometrial layer) were detected. A transcriptomic analysis on the uteri of young TG mice showed an up regulation of genes involved in cell cycle control and a down regulation of genes related to the immune system and the metabolism of xenobiotics. Aged TG females developed tumor masses in the uteri, which resembled an Endometrial Cancer (EC). Microarray and immunohistochemistry data indicated the deregulation of signaling pathways which are known to be altered in human ECs. The analysis of a cohort of 126 human ECs showed that LH-R overexpression is associated with early-stage tumors. Overall, our data led support to conclude that LH-R overexpression may directly contribute to trigger the neoplastic transformation of the endometrium.
- Published
- 2021
- Full Text
- View/download PDF
7. Evaluation of RAS Mutational Status in Liquid Biopsy to Monitor Disease Progression in Metastatic Colorectal Cancer Patients
- Author
-
Elena Lastraioli, Alessandra Bettiol, Jessica Iorio, Elvira Limatola, Daniele Checcacci, Erica Parisi, Cristina Bianchi, Annarosa Arcangeli, Mauro Iannopollo, Francesco Di Costanzo, and Marco Di Lieto
- Subjects
KRAS ,NRAS ,liquid biopsy ,metastatic colorectal cancer ,Cytology ,QH573-671 - Abstract
In this study we evaluated both~ K- and N-RAS mutations in plasma samples from patients with metastatic colorectal cancer by means of the BEAMing technology, and we assessed their diagnostic performance compared to RAS analyses performed on tissue. The sensitivity of BEAMing in identifying KRAS mutations was of 89.5%, with a fair specificity. The agreement with tissue analysis was moderate. The sensitivity for NRAS was high with a good specificity, and the agreement between tissue analysis and BEAMing was fair. Interestingly, significantly higher mutant allele fraction (MAF) levels were detected in patients with G2 tumors, liver metastases, and in those who did not receive surgery. NRAS MAF level was significantly higher in patients with mucinous adenocarcinoma and for those with lung metastases. A sharp increase in the MAF values was observed in patients who moved towards disease progression. More strikingly, molecular progression always anticipated the radiological one in these patients. These observations pave the way to the possibility of using liquid biopsy to monitor patients during treatment, and to enable oncologists to anticipate interventions compared to radiological analyses. This will allow time to be saved and ensure a better management of metastatic patients in the near future.
- Published
- 2023
- Full Text
- View/download PDF
8. Circulating miRNome profiling data in Behçet's syndrome
- Author
-
Giacomo Bagni, Giacomo Emmi, Elena Lastraioli, Francesca Di Patti, Elena Silvestri, Angela Guerriero, Serena Pillozzi, Elena Niccolai, Amedeo Amedei, Lorenzo Emmi, Domenico Prisco, and Annarosa Arcangeli
- Subjects
microRNA ,Circulating miRNAs ,Behçet ,Microarray ,Biomarker ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
We conducted a screening analysis to assess the presence of a characteristic extracellular circulating microRNAs (ci-miRNAs) profile in Behçet's syndrome (BS).Total RNA was extracted from platelets-free plasma (PFP) samples obtained from 16 BS patients and 18 healthy controls. Ci-miRNAs profiling was conducted by using dedicated Agilent microarray hybridization and data extraction technology. Statistical analysis of data extracted from microarray scanning revealed the deregulation of 36 ci-miRNAs, which turned out be differentially expressed between BS patients and healthy controls. Detailed experimental methods and data analysis were described here.The raw and normalized microarray data were deposited into Gene Expression Omnibus (GEO) under accession number GSE145191.
- Published
- 2021
- Full Text
- View/download PDF
9. Prognostic role of hERG1 Potassium Channels in Neuroendocrine Tumours of the Ileum and Pancreas
- Author
-
Jessica Iorio, Lorenzo Antonuzzo, Emanuela Scarpi, Massimo D’Amico, Claudia Duranti, Luca Messerini, Clotilde Sparano, Damiano Caputo, Daniele Lavacchi, Domenico Borzomati, Alice Antonelli, Lorenzo Nibid, Giuseppe Perrone, Alessandro Coppola, Roberto Coppola, Francesco di Costanzo, Elena Lastraioli, and Annarosa Arcangeli
- Subjects
hERG1 channel ,neuroendocrine tumours ,ileum ,pancreas ,prognosis ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
hERG1 potassium channels are widely expressed in human cancers of different origins, where they affect several key aspects of cellular behaviour. The present study was designed to evaluate the expression and clinical relevance of hERG1 protein in cancer tissues from patients suffering from neuroendocrine tumours (NETs) of ileal (iNETs) and pancreatic (pNETs) origin, with available clinicopathological history and follow-up. The study was carried out by immunohistochemistry with an anti-hERG1 monoclonal antibody. In a subset of samples, a different antibody directed against the hERG1/β1 integrin complex was also used. The analysis showed for the first time that hERG1 is expressed in human NETs originating from either the ileum or the pancreas. hERG1 turned out to have a prognostic value in NETs, showing (i) a statistically significant positive impact on OS of patients affected by ileal NETs, regardless the TNM stage; (ii) a statistically significant positive impact on OS of patients affected by aggressive (TNM stage IV) disease, either ileal or pancreatic; (iii) a trend to a negative impact on OS of patients affected by less aggressive (TNM stage I-III) disease, either ileal or pancreatic. Moreover, in order to evaluate whether ERG1 was functionally expressed in a cellular model of pNET, the INS1E rat insulinoma cell line was used, and it emerged that blocking ERG1 with a specific inhibitor of the channel (E4031) turned out in a significant reduction in cell proliferation.
- Published
- 2022
- Full Text
- View/download PDF
10. The Transcriptional Landscape of BRAF Wild Type Metastatic Melanoma: A Pilot Study
- Author
-
Elena Lastraioli, Federico Alessandro Ruffinatti, Giacomo Bagni, Luca Visentin, Francesco di Costanzo, Luca Munaron, and Annarosa Arcangeli
- Subjects
metastatic melanoma ,wild type BRAF ,transcriptomics ,microdissection ,ribosomes ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Melanoma is a relatively rare disease worldwide; nevertheless, it has a great relevance in some countries, such as in Europe. In order to shed some light upon the transcriptional profile of skin melanoma, we compared the gene expression of six independent tumours (all progressed towards metastatic disease and with wild type BRAF) to the expression profile of non-dysplastic melanocytes (considered as a healthy control) in a pilot study. Paraffin-embedded samples were manually micro-dissected to obtain enriched samples, and then, RNA was extracted and analysed through a microarray-based approach. An exhaustive bioinformatics analysis was performed to identify differentially expressed transcripts between the two groups, as well as enriched functional terms. Overall, 50 up- and 19 downregulated transcripts were found to be significantly changed in the tumour compared to the control tissue. Among the upregulated transcripts, the majority belonged to the immune response group and to the proteasome, while most of the downregulated genes were related to cytosolic ribosomes. A Gene Set Enrichment Analysis (GSEA), along with the RNA-Seq data retrieved from the TCGA/GTEx databases, confirmed the general trend of downregulation affecting cytoribosome proteins. In contrast, transcripts coding for mitoribosome proteins showed the opposite trend.
- Published
- 2022
- Full Text
- View/download PDF
11. KV11.1 Potassium Channel and the Na+/H+ Antiporter NHE1 Modulate Adhesion-Dependent Intracellular pH in Colorectal Cancer Cells
- Author
-
Jessica Iorio, Claudia Duranti, Tiziano Lottini, Elena Lastraioli, Giacomo Bagni, Andrea Becchetti, and Annarosa Arcangeli
- Subjects
hERG1 ,integrins ,Collagen I ,beta 1 integrin subunit ,cariporide ,lateral motility ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Increasing evidence indicates that ion channels and transporters cooperate in regulating different aspects of tumor pathophysiology. In cancer cells, H+/HCO3- transporters usually invert the transmembrane pH gradient typically observed in non-neoplastic cells, which is thought to contribute to cancer malignancy. To what extent the pH-regulating transporters are functionally linked to K+ channels, which are central regulators of cell membrane potential (Vm), is unclear. We thus investigated in colorectal cancer cells the implication of the pH-regulating transporters and KV11.1 (also known as hERG1) in the pH modifications stimulated by integrin-dependent cell adhesion. Colorectal cancer cell lines (HCT 116 and HT 29) were seeded onto β1 integrin-dependent substrates, collagen I and fibronectin. This led to a transient cytoplasmic alkalinization, which peaked at 90 min of incubation, lasted approximately 180 min, and was inhibited by antibodies blocking the β1 integrin. The effect was sensitive to amiloride (10 µM) and cariporide (5 µM), suggesting that it was mainly caused by the activity of the Na+/H+ antiporter NHE1. Blocking KV11.1 with E4031 shows that channel activity contributed to modulate the β1 integrin-dependent pHi increase. Interestingly, both NHE1 and KV11.1 modulated the colorectal cancer cell motility triggered by β1 integrin-dependent adhesion. Finally, the β1 integrin subunit, KV11.1 and NHE1 co-immunoprecipitated in colorectal cancer cells seeded onto Collagen I, suggesting the formation of a macromolecular complex following integrin-mediated adhesion. We conclude that the interaction between KV11.1, NHE1, and β1 integrin contributes to regulate colorectal cancer intracellular pH in relation to the tumor microenvironment, suggesting novel pharmacological targets to counteract pro-invasive and, hence, pro-metastatic behavior in colorectal cancer.
- Published
- 2020
- Full Text
- View/download PDF
12. Focus on Triple-Negative Breast Cancer: Potassium Channel Expression and Clinical Correlates
- Author
-
Elena Lastraioli
- Subjects
potassium channels ,breast cancer ,triple-negative breast cancer ,treatment ,prognosis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Despite improvements in early diagnosis and treatment, breast cancer is still a major health problem worldwide. Among breast cancer subtypes, the most challenging and harder to treat is represented by triple-negative molecular subtype. Due to its intrinsic features this subtype cannot be treated neither with hormonal therapy (since it does not express estrogen or progesterone receptors) nor with epidermal growth factor receptor 2 (HER2) inhibitors (as it does not express high levels of this protein). For these reasons, the standard of care for these patients is represented by a combination of surgery, radiation therapy and chemotherapy. In this scenario, searching for novel biomarkers that might help both in diagnosis and therapy is mandatory. In the last years, it was shown that different families of potassium channels are overexpressed in primary breast cancers. The altered ion channel expression may be useful for diagnostic and therapeutic purposes due to some peculiar characteristics of this class of molecules. Ion channels are defined as pore-forming transmembrane proteins regulating passive ion fluxes in the cells. Ion channels represent good potential markers since, being localized at the plasma membrane level, their detection and block with specific drugs and antibodies might be fast and tunable. This review focuses on triple-negative breast cancers and recapitulates the current knowledge about potassium channels' clinical relevance and their potential use in the clinical setting, for triple-negative breast cancer diagnosis and therapy.
- Published
- 2020
- Full Text
- View/download PDF
13. hERG1 and HIF-2α Behave as Biomarkers of Positive Response to Bevacizumab in Metastatic Colorectal Cancer Patients
- Author
-
Jessica Iorio, Elena Lastraioli, Lorenzo Tofani, Giulia Petroni, Lorenzo Antonuzzo, Luca Messerini, Giuseppe Perrone, Damiano Caputo, Maria Francesconi, Maria Michelina Amato, Moris Cadei, Giuseppina Arcangeli, Vincenzo Villanacci, Luca Boni, Roberto Coppola, Francesco Di Costanzo, and Annarosa Arcangeli
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: In search of novel biomarkers of response to bevacizumab in metastatic colorectal cancer (mCRC), we analyzed the expression and prognostic role of several proteins related to angiogenesis. Methods: A retrospective, multicenter study on 80 surgical samples from mCRC patients treated in first line with bevacizumab plus chemotherapy was accomplished. The following proteins were analyzed by immunohistochemistry: hERG1 potassium channel, β1-integrin, pAKT, NFkB, HIF-1α, HIF-2α, p53, VEGF-A, GLUT-1, and CA-IX. Data were analyzed in conjunction with the clinicopathological characteristics of the patients, KRAS status, response to bevacizumab, and follow-up. Results: (1) All the proteins were expressed in the samples, with statistically significant associations between HIF-1α and gender, HIF-2α and left colon, hERG1 and VEGF-A, β1-integrin and HIF-2α, GLUT-1 and both HIF-1α and HIF-2α, and CA-IX and VEGF-A. (2) At the univariate analysis, positivity for hERG1, VEGF-A, and the active form of HIF-2α (aHIF-2α), and the G3 histological grade showed a positive impact on progression-free survival (PFS). (3) hERG1 and aHIF-2α maintained their positive impact on PFS at the multivariate analysis. (4) hERG1 behaved as a protective factor for PFS independently on KRAS status. Conclusions: hERG1 and aHIF-2α might help to identify patients who would benefit from bevacizumab treatment.
- Published
- 2020
- Full Text
- View/download PDF
14. hERG1 channel expression associates with molecular subtypes and prognosis in breast cancer
- Author
-
Jessica Iorio, Icro Meattini, Simonetta Bianchi, Marco Bernini, Virginia Maragna, Luca Dominici, Donato Casella, Vania Vezzosi, Lorenzo Orzalesi, Jacopo Nori, Lorenzo Livi, Annarosa Arcangeli, and Elena Lastraioli
- Subjects
hERG1 ,Potassium channels ,Breast cancer ,Molecular subtype ,Immunohistochemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background Breast cancer (BC) is the most frequent malignancy among females worldwide. Despite several efforts and improvements in early diagnosis and treatment, there are still tumors characterized by an aggressive behavior due to unfavorable biology, thus quite difficult to treat. In this view, searching for novel potential biomarkers is mandatory. Among them, in the recent years data have been gathered addressing ion channel as important players in oncology. Methods A retrospective pilot study was performed on 40 BC samples by means of immunohistochemistry in order to evaluate hERG1 potassium channels expression in BC. Results We provide evidence that hERG1 is expressed in all the BC samples analyzed. hERG1 expression was significantly associated with molecular subtype with the highest expression in Luminal A and the lowest in basal-like tumors (p = 0.001), tumor grading (the highest hERG1 expression in well-moderate differentiated tumors, p = 0.020), estrogen receptors (high hERG1 expression in ER-positive samples, p = 0.008) and Ki67 proliferative index (high hERG1 scoring in samples with low proliferative index, p = 0.038). Also, a p value close to significance was noticed for the association between hERG1 and HER2 expression (p = 0.079). At the survival analysis, patients with high hERG1 expression turned out to have a longer progression-free survival, although statistical significance was not reached (p = 0.195). The same trend was observed analyzing local relapse free-survival (LRFS) and metastases-free survival (MFS): patients with higher hERG1 scoring had longer LRFS and MFS (p = 0.124 and p = 0.071, respectively). Conclusions The results of this pilot study provide the first evidence that the hERG1 protein is expressed in primary BC, and its expression associates with molecular subtype. hERG1 apparently behaves as a protective factor, since it contributes to identify a subset of patients with better outcome. Overall, these data suggest that hERG1 might be an additional tool for the management of BC, nevertheless further investigations are warranted to better clarify hERG1 role and clinical usefulness in BC.
- Published
- 2018
- Full Text
- View/download PDF
15. Circulating Endothelial Progenitor Cells in Type 1 Diabetic Patients: Relation with Patients’ Age and Disease Duration
- Author
-
Adolfo Arcangeli, Elena Lastraioli, Barbara Piccini, Massimo D’Amico, Lorenzo Lenzi, Serena Pillozzi, Maria Calabrese, Sonia Toni, and Annarosa Arcangeli
- Subjects
type 1 diabetes mellitus ,endothelial progenitor cells ,flow cytometry ,diabetes duration ,patients’ age ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
ObjectivesCirculating endothelial progenitor cells (cEPCs) have been reported to be dysfunctional in diabetes mellitus (DM) patients, accounting for the vascular damage and the ensuing high risk for cardiovascular disease (CVD) characteristic of this disease. The aim of the present study was to evaluate the number of circulating cEPCs in type 1 DM (T1DM) patients, without clinical vascular damage, of different ages and with different disease duration.MethodsAn observational, clinical-based prospective study was performed on T1DM patients enrolled in two clinical centers. cEPCs were determined by flow cytometry, determining the number of CD34/CD133/VEGFR2-positive cells within peripheral blood mononuclear cells (PBMCs).ResultsThe number of cEPCs was lower in adult T1DM patients, whilst higher in childhood/young patients, compared to controls of the same age range. When patients were grouped into two age groups (≥ or
- Published
- 2017
- Full Text
- View/download PDF
16. Immunohistochemical Biomarkers in Gastric Cancer Research and Management
- Author
-
Elena Lastraioli, Maria Raffaella Romoli, and Annarosa Arcangeli
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Gastric cancer still represents a major health problem, despite a decrease in its incidence in the last years. Due to the social impact of gastric cancer (GC), there is a need for novel biomarkers in order to stratify patients into appropriate screening, surveillance, or treatment programs. Although histopathology remains the most reliable and less expensive method, numerous efforts have been made searching for novel biomarkers. In recent years, several molecules have been identified and tested for their clinical relevance in GC management. In this paper, we will focus on a well-known GC marker, whose determination is mandatory in GC, HER2, a marker whose correlation with prognosis is still controversial (VEGF-A) and a quite novel, unconventional marker, the ether-à-go-go-related gene 1 (hERG1). All these proteins can be easily detected with immunohistochemistry, a technique widely used both in diagnostic and research laboratories that represents a link between surgical and molecular pathology, basic science, and clinical medicine.
- Published
- 2012
- Full Text
- View/download PDF
17. Combination Therapy with a Bispecific Antibody Targeting the hERG1/β1 Integrin Complex and Gemcitabine in Pancreatic Ductal Adenocarcinoma
- Author
-
Tiziano Lottini, Claudia Duranti, Jessica Iorio, Michele Martinelli, Rossella Colasurdo, Franco Nicolás D’Alessandro, Matteo Buonamici, Stefano Coppola, Valentina Devescovi, Vincenzo La Vaccara, Alessandro Coppola, Roberto Coppola, Elena Lastraioli, and Annarosa Arcangeli
- Subjects
Cancer Research ,Oncology ,PDAC ,K+ channels ,engineered antibodies ,xenograft ,ultrasound ,photoacoustic imaging - Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents an unmet medical need. Difficult/late diagnosis as well as the poor efficacy and high toxicity of chemotherapeutic drugs result in dismal prognosis. With the aim of improving the treatment outcome of PDAC, we tested the effect of combining Gemcitabine with a novel single chain bispecific antibody (scDb) targeting the cancer-specific hERG1/β1 integrin complex. First, using the scDb (scDb-hERG1-β1) in immunohistochemistry (IHC), Western blot (WB) analysis and immunofluorescence (IF), we confirmed the presence of the hERG1/β1 integrin complex in primary PDAC samples and PDAC cell lines. Combining Gemcitabine with scDb-hERG1-β1 improved its cytotoxicity on all PDAC cells tested in vitro. We also tested the combination treatment in vivo, using an orthotopic xenograft mouse model involving ultrasound-guided injection of PDAC cells. We first demonstrated good penetration of the scDb-hERG1-β1 conjugated with indocyanine green (ICG) into tumour masses by photoacoustic (PA) imaging. Next, we tested the effects of the combination at either therapeutic or sub-optimal doses of Gemcitabine (25 or 5 mg/kg, respectively). The combination of scDb-hERG1-β1 and sub-optimal doses of Gemcitabine reduced the tumour masses to the same extent as the therapeutic doses of Gemcitabine administrated alone; yielded increased survival; and was accompanied by minimised side effects (toxicity). These data pave the way for a novel therapeutic approach to PDAC, based on the combination of low doses of a chemotherapeutic drug (to minimize adverse side effects and the onset of resistance) and the novel scDb-hERG1-β1 targeting the hERG1/β1 integrin complex as neoantigen.
- Published
- 2023
- Full Text
- View/download PDF
18. Potassium in Solid Cancers
- Author
-
Lisa Lastraioli, Elena Lastraioli, and Jessica Iorio
- Subjects
chemistry ,business.industry ,Potassium ,Medicine ,chemistry.chemical_element ,business ,Nuclear chemistry - Abstract
Electrolyte disorders are a frequent finding in cancer patients. In the majority of cases the etiologies of such disorders are common to all cancer types (i.e. diuretic-induced hyponatremia or hypokalemia). Sometimes, electrolyte disorders are caused by paraneoplastic syndromes or are due to cancer therapy. Potassium is one of the most important electrolytes of the human body since it is involved in the regulation of muscle contraction, maintenance of the integrity of the skeleton, blood pressure and nerve transmission as well as in the normal function of cells. Potassium homeostasis is strictly regulated since the gap between the recommended daily dietary intake (120 mEq/day) and the levels stored in the extracellular fluid (around 70 mEq) is huge. Alterations of potassium homeostasis are frequent in cancer patients as well alterations in potassium channels, the transmembrane proteins that mediate potassium fluxes within the cells. The present chapter is focused on the clinical significance of potassium homeostasis and potassium channels in patients with solid tumors.
- Published
- 2021
19. Circulating miRNome profiling data in Behçet's syndrome
- Author
-
Annarosa Arcangeli, Lorenzo Emmi, Domenico Prisco, Francesca Di Patti, Amedeo Amedei, Elena Lastraioli, Serena Pillozzi, Angela Guerriero, Elena Silvestri, Elena Niccolai, Giacomo Emmi, and Giacomo Bagni
- Subjects
Gene expression omnibus ,Multidisciplinary ,S syndrome ,Science (General) ,Microarray ,microRNA ,Behçet ,Microarray analysis techniques ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Computational biology ,Biomarker ,Biology ,Biomarker (cell) ,Circulating MicroRNA ,Q1-390 ,Statistical analysis ,Experimental methods ,Circulating miRNAs ,Data Article - Abstract
We conducted a screening analysis to assess the presence of a characteristic extracellular circulating microRNAs (ci-miRNAs) profile in Behcet's syndrome (BS). Total RNA was extracted from platelets-free plasma (PFP) samples obtained from 16 BS patients and 18 healthy controls. Ci-miRNAs profiling was conducted by using dedicated Agilent microarray hybridization and data extraction technology. Statistical analysis of data extracted from microarray scanning revealed the deregulation of 36 ci-miRNAs, which turned out be differentially expressed between BS patients and healthy controls. Detailed experimental methods and data analysis were described here. The raw and normalized microarray data were deposited into Gene Expression Omnibus (GEO) under accession number {"type":"entrez-geo","attrs":{"text":"GSE145191","term_id":"145191"}}GSE145191.
- Published
- 2021
20. A Transcriptomic Approach Reveals Selective Ribosomal Remodelling in the Tumour Versus the Stromal Compartment of Metastatic Colorectal Cancer
- Author
-
Cesare Sala, Luca Munaron, Francesco Di Costanzo, Federico Alessandro Ruffinatti, Elena Lastraioli, and Annarosa Arcangeli
- Subjects
Cancer Research ,Stromal cell ,Microarray ,Angiogenesis ,colorectal cancer ,Biology ,Article ,ribosomes ,Transcriptome ,03 medical and health sciences ,transcriptomics ,0302 clinical medicine ,Ribosomal protein ,microdissection ,stroma ,Gene expression ,Eukaryotic Small Ribosomal Subunit ,Gene ,RC254-282 ,030304 developmental biology ,0303 health sciences ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,Cancer research - Abstract
Because of its high incidence and poor prognosis, colorectal cancer (CRC) represents an important health issue in several countries. As with other carcinomas, the so-called tumour microenvironment (TME) has been shown to play key roles in CRC progression and related therapeutical outcomes, even though a deeper understanding of the underlying molecular mechanisms is needed to devise new treatment strategies. For some years now, omics technologies and consolidated bioinformatics pipelines have allowed scientists to access large amounts of biologically relevant information, even when starting from small tissue samples, thus, in order to shed new light upon the role of the TME in CRC, we compared the gene expression profiles of 6 independent tumour tissues (all progressed towards metastatic disease) to the expression profile of the surrounding stromata. To do this, paraffin-embedded whole tissues were first microdissected to obtain samples enriched with tumour and stromal cells, respectively. Afterwards, RNA was extracted and analysed using a microarray-based approach. A thorough bioinformatics analysis was then carried out to identify transcripts differentially expressed between the two groups and possibly enriched functional terms. Overall, 193 genes were found to be significantly downregulated in tumours compared to the paired stromata. The functional analysis of the downregulated gene list revealed three principal macro areas of interest: the extracellular matrix, cell migration, and angiogenesis. Conversely, among the upregulated genes, the main alterations detected by the functional annotation were related to the ribosomal proteins (rProteins) of both the large (60S) and small (40S) subunits of the cytosolic ribosomes. Subsequent gene set enrichment analysis (GSEA) confirmed the massive overexpression of most cytosolic—but not mitochondrial—ribosome rProteins.
- Published
- 2021
- Full Text
- View/download PDF
21. Neonatal Nav1.5 Protein Expression in Human Colorectal Cancer: Immunohistochemical Characterization and Clinical Evaluation
- Author
-
Giuseppe Perrone, Giulia Cerino, Emanuela Scarpi, Vincenzo Villanacci, Lapo Bencini, Niccolo' Ghezzi, Luca Messerini, R. Mine Guzel, Scott P. Fraser, Jessica Iorio, Elena Lastraioli, Mustafa B.A. Djamgoz, and Annarosa Arcangeli
- Subjects
HUMAN PROSTATE-CANCER ,0301 basic medicine ,Cancer Research ,Colorectal cancer ,INVASION ,colorectal cancer ,Article ,Metastasis ,03 medical and health sciences ,Basal (phylogenetics) ,0302 clinical medicine ,voltage-gated sodium channel ,Medicine ,metastasis ,1112 Oncology and Carcinogenesis ,Clinical significance ,neonatal Nav1.5 ,immunohistochemistry ,RC254-282 ,Univariate analysis ,Science & Technology ,biology ,business.industry ,COLON-CANCER ,GLUT-1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,neonatal Nav1 ,STAGE-I ,030104 developmental biology ,Oncology ,SODIUM-CHANNEL EXPRESSION ,MARKER ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,GROWTH ,Immunohistochemistry ,GLUT1 ,business ,Life Sciences & Biomedicine ,Companion diagnostic - Abstract
Simple Summary The voltage-gated sodium channel is a type of protein normally expressed in the ‘excitable’ tissues (nerves and muscles) of the body. Epithelial tissues (gut, lungs etc.), which are normally devoid of such a channel, express it at high levels upon becoming cancerous. This occurs also in colorectal cancer cells where the channel subtype is the embryonic (‘neonatal’) variant, nNav1.5. In colorectal cancer cells, as in other solid cancer cells, channel activity promotes invasiveness. However, there is little information on the status of nNav1.5 in human colorectal tissues and how this might relate to patient outcome. Here, we show (i) that nNav1.5 expression is much higher in cancer tissues compared with normal; (ii) that nNav1.5 co-occurs with several other biomarkers of pathological importance; and (iii) that disease-free survival of colorectal patients is inversely correlated with channel expression. In conclusion, nNav1.5 has combined diagnostic and therapeutic potential in clinical management of colorectal cancer. Abstract Voltage-gated Na+ channels (VGSCs) are expressed widely in human carcinomas and play a significant role in promoting cellular invasiveness and metastasis. However, human tissue-based studies and clinical characterization are lacking. In several carcinomas, including colorectal cancer (CRCa), the predominant VGSC is the neonatal splice variant of Nav1.5 (nNav1.5). The present study was designed to determine the expression patterns and clinical relevance of nNav1.5 protein in human CRCa tissues from patients with available clinicopathological history. The immunohistochemistry was made possible by the use of a polyclonal antibody (NESOpAb) specific for nNav1.5. The analysis showed that, compared with normal mucosa, nNav1.5 expression occurred in CRCa samples (i) at levels that were significantly higher and (ii) with a pattern that was more delineated (i.e., apical/basal or mixed). A surprisingly high level of nNav1.5 protein expression also occurred in adenomas, but this was mainly intracellular and diffuse. nNav1.5 showed a statistically significant association with TNM stage, highest expression being associated with TNM IV and metastatic status. Interestingly, nNav1.5 expression co-occurred with other biomarkers associated with metastasis, including hERG1, KCa3.1, VEGF-A, Glut1, and EGFR. Finally, univariate analysis showed that nNav1.5 expression had an impact on progression-free survival. We conclude (i) that nNav1.5 could represent a novel clinical biomarker (‘companion diagnostic’) useful to better stratify CRCa patients and (ii) that since nNav1.5 expression is functional, it could form the basis of anti-metastatic therapies including in combination with standard treatments.
- Published
- 2021
- Full Text
- View/download PDF
22. A unique circulating miRNA profile highlights thrombo-inflammation in Behçet’s syndrome
- Author
-
Giacomo Bagni, Elena Lastraioli, Annarosa Arcangeli, Maria Letizia Urban, Alessandra Bettiol, Lorenzo Emmi, Domenico Prisco, Claudia Fiorillo, Elena Silvestri, Francesca Di Patti, Giacomo Emmi, and Matteo Becatti
- Subjects
Adult ,Male ,Oncology ,Behçet Syndrome ,medicine.medical_specialty ,Microarray ,Giant Cell Arteritis ,Immunology ,Inflammation ,medicine.disease_cause ,Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Rheumatology ,cardiovascular disease ,Internal medicine ,medicine ,Humans ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,autoimmune diseases ,Circulating MicroRNA ,Prospective Studies ,Thromboinflammation ,Receiver operating characteristic ,business.industry ,Behcet Syndrome ,Gene Expression Profiling ,medicine.disease ,MicroRNAs ,Giant cell arteritis ,ROC Curve ,inflammation ,Case-Control Studies ,Cohort ,Biomarker (medicine) ,Female ,medicine.symptom ,systemic vasculitis ,business ,Biomarkers ,Oxidative stress ,Systemic vasculitis - Abstract
ObjectivesBehçet’s syndrome (BS) is a rare systemic vasculitis often complicated by thrombotic events. Given the lack of validated biomarkers, BS diagnosis relies on clinical criteria.In search of novel biomarkers for BS diagnosis, we determined the profile of plasmatic circulating microRNAs (ci-miRNAs) in patients with BS compared with healthy controls (HCs).Methodsci-miRNA profile was evaluated by microarray in a screening cohort (16 patients with BS and 18 HCs) and then validated by poly(T) adaptor PCR (PTA-PCR) in a validation cohort (30 patients with BS and 30 HCs). Two disease control groups (30 patients with systemic lupus erythematosus (SLE) and 30 patients with giant cell arteritis (GCA) were also analysed.ResultsFrom the microarray screening, 29 deregulated (differentially expressed (DE)) human ci-miRNAs emerged. A hierarchical cluster analysis indicated that DE ci-miRNAs clearly segregated patients from controls, independently of clinical features. PTA-PCR analysis on the validation cohort confirmed the deregulation of miR-224-5p, miR-206 and miR-653-5p. The combined receiver operating characteristic (ROC) curve analyses showed that such ci-miRNAs discriminate BS from HCs (and BS with active vs inactive disease), as well as BS from patients with SLE and GCA.The functional annotation analyses (FAAs) showed that the most enriched pathways affected by DE ci-miRNAs (ie, cell–matrix interaction, oxidative stress and blood coagulation) are related to thrombo-inflammatory mechanisms. Accordingly, the expression of the three ci-miRNAs from the validation cohort significantly correlated with leucocyte reactive oxygen species production and plasma lipid peroxidation.ConclusionsThe ci-miRNA profile identified in this study may represent a novel, poorly invasive BS biomarker, while suggesting an epigenetic control of BS-related thrombo-inflammation.
- Published
- 2021
23. Transgenic mice overexpressing the LH receptor in the female reproductive system spontaneously develop endometrial tumour masses
- Author
-
Annarosa Arcangeli, Elena Lastraioli, Cesare Sala, Ivo Noci, Miriam Armenio, Laura Carraresi, Tiziano Lottini, Serena Pillozzi, Claudia Duranti, and Jessica Iorio
- Subjects
Genetically modified mouse ,Cell biology ,LH receptor, endometrial cancer, transgenic mice ,Science ,Transgene ,Uterus ,Down-Regulation ,Mice, Transgenic ,Diseases ,Ovary ,Pathogenesis ,Biology ,Endometrium ,Article ,Cohort Studies ,Mice ,medicine ,Animals ,Humans ,Neoplastic transformation ,Cancer ,Multidisciplinary ,Endometrial cancer ,Biological techniques ,luteinizing hormone/choriogonadotropin receptor ,Genitalia, Female ,Receptors, LH ,medicine.disease ,Endometrial Neoplasms ,Up-Regulation ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Risk factors ,Cancer research ,Medicine ,Female ,Transcriptome ,Biomarkers - Abstract
The receptor for the luteinizing hormone (LH-R) is aberrantly over expressed in cancers of the reproductive system. To uncover whether LH-R over expression has a causative role in cancer, we generated a transgenic (TG) mouse which overexpresses the human LH-R (hLH-R) in the female reproductive tract, under the control of the oviduct-specific glycoprotein (OGP) mouse promoter (mogp-1). The transgene was highly expressed in the uterus, ovary and liver, but only in the uterus morphological and molecular alterations (increased proliferation and trans-differentiation in the endometrial layer) were detected. A transcriptomic analysis on the uteri of young TG mice showed an up regulation of genes involved in cell cycle control and a down regulation of genes related to the immune system and the metabolism of xenobiotics. Aged TG females developed tumor masses in the uteri, which resembled an Endometrial Cancer (EC). Microarray and immunohistochemistry data indicated the deregulation of signaling pathways which are known to be altered in human ECs. The analysis of a cohort of 126 human ECs showed that LH-R overexpression is associated with early-stage tumors. Overall, our data led support to conclude that LH-R overexpression may directly contribute to trigger the neoplastic transformation of the endometrium.
- Published
- 2021
24. KV11.1 Potassium Channel and the Na+/H+ Antiporter NHE1 Modulate Adhesion-Dependent Intracellular pH in Colorectal Cancer Cells
- Author
-
Tiziano Lottini, Claudia Duranti, Giacomo Bagni, Andrea Becchetti, Jessica Iorio, Annarosa Arcangeli, Elena Lastraioli, Iorio, J, Duranti, C, Lottini, T, Lastraioli, E, Bagni, G, Becchetti, A, and Arcangeli, A
- Subjects
0301 basic medicine ,integrin ,Antiporter ,Intracellular pH ,Integrin ,lateral motility ,03 medical and health sciences ,0302 clinical medicine ,beta 1 integrin subunit ,medicine ,Pharmacology (medical) ,Cell adhesion ,Ion channel ,Collagen I ,cariporide ,hERG1 ,integrins ,Pharmacology ,biology ,Chemistry ,lcsh:RM1-950 ,Cancer ,medicine.disease ,Potassium channel ,Cell biology ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein - Abstract
Increasing evidence indicates that ion channels and transporters cooperate in regulating different aspects of tumor pathophysiology. In cancer cells, H+/HCO3- transporters usually invert the transmembrane pH gradient typically observed in non-neoplastic cells, which is thought to contribute to cancer malignancy. To what extent the pH-regulating transporters are functionally linked to K+ channels, which are central regulators of cell membrane potential (Vm), is unclear. We thus investigated in colorectal cancer cells the implication of the pH-regulating transporters and KV11.1 (also known as hERG1) in the pH modifications stimulated by integrin-dependent cell adhesion. Colorectal cancer cell lines (HCT 116 and HT 29) were seeded onto 1 integrin-dependent substrates, collagen I and fibronectin. This led to a transient cytoplasmic alkalinization, which peaked at 90 minutes of incubation, lasted approximately 180 min, and was inhibited by antibodies blocking the 1 integrin. The effect was sensitive to amiloride (10 µM) and cariporide (5 µM), suggesting that it was mainly caused by the activity of the Na+/H+ antiporter NHE1. Blocking KV11.1 with E4031 shows that channel activity contributed to modulate the 1 integrin-dependent pHi increase. Interestingly, both NHE1 and KV11.1 modulated the colorectal cancer cell motility triggered by 1 integrin-dependent adhesion. Finally, the 1 integrin subunit, KV11.1 and NHE1 co-immunoprecipitated in colorectal cancer cells seeded onto Collagen I, suggesting the formation of a macromolecular complex following integrin-mediated adhesion. We conclude that the interaction between KV11.1, NHE1 and β1 integrin contribute to regulate colorectal cancer intracellular pH in relation to the tumor microenvironment, suggesting novel pharmacological targets to counteract pro-invasive and, hence, pro-metastatic behavior in colorectal cancer.
- Published
- 2020
- Full Text
- View/download PDF
25. Evaluation of RAS mutational status through BEAMing assay to monitor disease progression of metastatic colorectal cancer: a case report
- Author
-
Elena Lastraioli, Valeria Emma Palmieri, Lorenzo Antonuzzo, Luca Messerini, Daniele Lavacchi, Francesca Castiglione, and Francesco Di Costanzo
- Subjects
0301 basic medicine ,Oncology ,FOLFIRI-Cetuximab Regimen ,Male ,Cancer Research ,Pyrrolidines ,Pyridines ,DNA Mutational Analysis ,Leucovorin ,Cetuximab ,medicine.disease_cause ,Trifluridine ,0302 clinical medicine ,Maintenance therapy ,FOLFOX ,Antineoplastic Combined Chemotherapy Protocols ,Pharmacology (medical) ,Middle Aged ,Bevacizumab ,Drug Combinations ,030220 oncology & carcinogenesis ,FOLFIRI ,Disease Progression ,KRAS ,Fluorouracil ,Colorectal Neoplasms ,medicine.drug ,medicine.medical_specialty ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Liquid biopsy ,neoplasms ,Capecitabine ,Pharmacology ,business.industry ,Phenylurea Compounds ,liquid biopsy, metastatic colorectal cancer, OncoBEAM, RAS mutation ,Liquid Biopsy ,digestive system diseases ,030104 developmental biology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Mutation ,ras Proteins ,Camptothecin ,business ,Thymine - Abstract
Since the introduction of antiepidermal growth factor receptor (anti-EGFR) monoclonal antibodies (moAbs), the treatment of metastatic colorectal cancer (mCRC) has become crucially dependent on the mutation profile of the tumour over the last two decades. Recently, rechallenge strategy with cetuximab-based chemotherapy has demonstrated to be active in a subgroup of patients whose tumour maintained wild-type RAS and RAF status. In this setting, liquid biopsy may replace tissue sample for the identification of specific subgroups of pretreated patients that may benefit from the reintroduction of anti-EGFR moAbs. In November 2014, a 64-year-old man with IVB stage BRAF, KRAS and NRAS wild-type mCRC was admitted in our hospital. He received FOLFIRI cetuximab as first-line treatment with deep and long-lasting partial response (PR), followed by cetuximab maintenance therapy until January 2016. At the time of disease progression, FOLFIRI cetuximab regimen was reintroduced resulting in stabilization of disease and he continued with capecitabine cetuximab therapy until disease progression in October 2016. Then, the patient consecutively received FOLFOX bevacizumab, TAS-102, regorafenib and FOLFIRI followed by de Gramont maintenance treatment. Finally, he was retreated with FOLFIRI cetuximab with disease progression within 3 months and died in May 2019. During his clinical course, liquid biopsy detected two mutations: one in KRAS Cd.12 and one in NRAS Cd. 61. The longitudinal assessment of RAS status offers considerable advantages in order to avoid side effects and economic costs for ineffective treatment choices. Liquid biopsy could help better monitor the disease and provide molecularly guided treatments.
- Published
- 2020
26. Transcriptomic data of bevacizumab-adapted colorectal adenocarcinoma cells HCT-116
- Author
-
Cesare, Sala, Tiziano, Lottini, Elena, Lastraioli, Alessandro, Ruffinatti Federico, Luca, Visentin, and Annarosa, Arcangeli
- Published
- 2023
- Full Text
- View/download PDF
27. hERG1 behaves as biomarker of progression to adenocarcinoma in Barrett's esophagus and can be exploited for a novel endoscopic surveillance
- Author
-
Luca Messerini, Tiziano Lottini, Anna Tomezzoli, Maria Novella Ringressi, Marilena Fazi, Jessica Iorio, Giancarlo Freschi, Luca Saragoni, Luca Boni, Elena Lastraioli, Vincenzo Villanacci, Antonio Taddei, Laura Carraresi, Maria Bencivenga, Paolo Bechi, Roberta La Mendola, Marianna Salemme, Mariella Chiudinelli, Claudia Duranti, Carla Vindigni, Ilaria Manzi, Bruno Compagnoni, Giovanni de Manzoni, and Annarosa Arcangeli
- Subjects
0301 basic medicine ,Esophageal Neoplasms ,Barrett’s esophagus ,adenocarcinoma progression ,hERG1 ,optical imaging ,surveillance ,Mice ,0302 clinical medicine ,Single-Chain Variable Fragment Antibody ,Mice, Inbred BALB C ,Adenocarcinoma progression ,Barrett's esophagus ,Optical imaging ,Surveillance ,Oncology ,Prognosis ,humanities ,Esophageal dysplasia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Biomarker (medicine) ,Research Paper ,Diagnostic Imaging ,Risk ,medicine.medical_specialty ,Mice, Transgenic ,03 medical and health sciences ,Barrett Esophagus ,Esophagus ,medicine ,Animals ,Humans ,Metaplasia ,business.industry ,General surgery ,Precursor lesion ,Endoscopy ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,Surgery ,Disease Models, Animal ,030104 developmental biology ,Case-Control Studies ,business ,Biomarkers - Abstract
// Elena Lastraioli 1 , Tiziano Lottini 1 , Jessica Iorio 1 , Giancarlo Freschi 2 , Marilena Fazi 2 , Claudia Duranti 1 , Laura Carraresi 3 , Luca Messerini 1 , Antonio Taddei 2 , Maria Novella Ringressi 2 , Marianna Salemme 4 , Vincenzo Villanacci 4 , Carla Vindigni 5 , Anna Tomezzoli 6 , Roberta La Mendola 7 , Maria Bencivenga 7 , Bruno Compagnoni 8 , Mariella Chiudinelli 9 , Luca Saragoni 10 , Ilaria Manzi 11 , Giovanni De Manzoni 7 , Paolo Bechi 2 , Luca Boni 12, * , Annarosa Arcangeli 1, * 1 Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy 2 Department of Surgery and Translational Medicine, University of Florence, 50134 Florence, Italy 3 DI.V.A.L Toscana Srl, 50019 Sesto Fiorentino, Italy 4 Institute of Pathology, Spedali Civili, 25123 Brescia, Italy 5 Pathology Division, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy 6 Pathology Division, Borgo Trento Hospital, 37134 Verona, Italy 7 Division of Surgery, University of Verona, 37134 Verona, Italy 8 Surgery Division, Esine Hospital, ASL Vallecamonica Sebino, 25040 Esine (BS), Italy 9 Pathology Division, Esine Hospital, ASL Vallecamonica Sebino, 25040 Esine (BS), Italy 10 Pathology Division, Morgagni-Pierantoni Hospital, 47121 Forli, Italy 11 Gastroenterology and Endoscopy Unit, Morgagni-Pierantoni Hospital, 47121 Forli, Italy 12 Clinical Trials Coordinating Center, Azienda Ospedaliero-Universitaria Careggi/Istituto Toscano Tumori, 50134 Florence, Italy * These authors contributed equally to this work Correspondence to: Annarosa Arcangeli, email: annarosa.arcangeli@unifi.it Keywords: hERG1, Barrett’s esophagus, adenocarcinoma progression, surveillance, optical imaging Received: March 13, 2016 Accepted: July 09, 2016 Published: August 09, 2016 ABSTRACT Barrett’s esophagus (BE) is the only well-known precursor lesion of esophageal adenocarcinoma (EA). The exact estimates of the annual progression rate from BE to EA vary from 0.07% to 3.6%. The identification of BE patients at higher risk to progress to EA is hence mandatory, although difficult to accomplish. In search of novel BE biomarkers we analyzed the efficacy of hERG1 potassium channels in predicting BE progression to EA. Once tested by immunohistochemistry (IHC) on bioptic samples, hERG1 was expressed in BE, and its expression levels increased during progression from BE to esophageal dysplasia (ED) and EA. hERG1 was also over-expressed in the metaplastic cells arising in BE lesions obtained in different BE mouse models, induced either surgically or chemically. Furthermore, transgenic mice which over express hERG1 in the whole gastrointestinal tract, developed BE lesions after an esophago-jejunal anastomosis more frequently, compared to controls. A case-control study was performed on 104 bioptic samples from newly diagnosed BE patients further followed up for at least 10 years. It emerged a statistically significant association between hERG1 expression status and risk of progression to EA. Finally, a novel fluorophore- conjugated recombinant single chain variable fragment antibody (scFv-hERG1-Alexa488) was tested on freshly collected live BE biopsies: it could recognize hERG1 positive samples, perfectly matching IHC data. Overall, hERG1 can be considered a novel BE biomarker to be exploited for a novel endoscopic surveillance protocol, either in biopsies or through endoscopy, to identify those BE patients with higher risk to progress to EA.
- Published
- 2016
28. hERG1 Potassium Channels: Novel Biomarkers in Human Solid Cancers
- Author
-
Tiziano Lottini, Lapo Bencini, Marco Bernini, Elena Lastraioli, and Annarosa Arcangeli
- Subjects
ERG1 Potassium Channel ,lcsh:Medicine ,Translational research ,Ovary ,Review Article ,Biology ,Endometrium ,General Biochemistry, Genetics and Molecular Biology ,Translational Research, Biomedical ,Mice ,In vivo ,Neoplasms ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,herg potassium channels, solid cancers ,Early Detection of Cancer ,Cell Proliferation ,Regulation of gene expression ,Cardiotoxicity ,General Immunology and Microbiology ,lcsh:R ,General Medicine ,Ether-A-Go-Go Potassium Channels ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Tumor progression ,Immunology ,Cancer research ,Pancreas - Abstract
Because of their high incidence and mortality solid cancers are a major health problem worldwide. Although several new biomarkers and potential targets for therapy have been identified through biomolecular research in the last years, the effects on patients’ outcome are still unsatisfactory. Increasing evidence indicates that hERG1 potassium channels are overexpressed in human primary cancers of different origin and several associations between hERG1 expression and clinicopathological features and/or outcome are emerging. Aberrant hERG1 expression may be exploited either for early diagnosis (especially in those cancers where it is expressed in the initial steps of tumor progression) or for therapy purposes. Indeed, hERG1 blockage impairs tumor cell growth bothin vitroandin vivoin preclinical mouse model. hERG1-based tumor therapy in humans, however, encounters the major hindrance of the potential cardiotoxicity that many hERG1 blockers exert. In this review we focus on recent advances in translational research in some of the most frequent human solid cancers (breast, endometrium, ovary, pancreas, esophagus, stomach, and colorectum) that have been shown to express hERG1 and that are a major health problem.
- Published
- 2015
29. hERG1 channels regulate VEGF-A secretion in human gastric cancer: clinicopathological correlations and therapeutical implications
- Author
-
Olivia, Crociani, Elena, Lastraioli, Luca, Boni, Serena, Pillozzi, Maria Raffaella, Romoli, Massimo, D'Amico, Matteo, Stefanini, Silvia, Crescioli, Alessio, Masi, Antonio, Taddei, Lapo, Bencini, Marco, Bernini, Marco, Farsi, Stefania, Beghelli, Aldo, Scarpa, Luca, Messerini, Anna, Tomezzoli, Carla, Vindigni, Paolo, Morgagni, Luca, Saragoni, Elisa, Giommoni, Silvia, Gasperoni, Francesco, Di Costanzo, Franco, Roviello, Giovanni, De Manzoni, Paolo, Bechi, and Annarosa, Arcangeli
- Subjects
hERG1 Channels ,VEGF-A Secretion ,GASTRIC CANCER ,Vascular Endothelial Growth Factor A ,ERG1 Potassium Channel ,Pathology ,medicine.medical_specialty ,Cancer Research ,Nude ,Mice, Nude ,Enzyme-Linked Immunosorbent Assay ,Biology ,Adenocarcinoma ,bevacizumab ,Real-Time Polymerase Chain Reaction ,Transfection ,Cell Line ,1 [2 (6 methyl 2 pyridyl)ethyl] 4 (4 methylsulfonylaminobenzoyl)piperidine ,Mice ,In vivo ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Secretion ,Grading (tumors) ,Ether-A-Go-Go Potassium Channels ,Heterografts ,Immunohistochemistry ,Reverse Transcriptase Polymerase Chain Reaction ,Oncology ,Medicine (all) ,Tumor ,bevacizumab, potassium channel HERG, potassium channel HERG1, unclassified drug, vasculotropin A ,medicine.disease ,potassium channel HERG1 ,potassium channel HERG ,unclassified drug ,Vascular endothelial growth factor A ,Real-time polymerase chain reaction ,vasculotropin A ,Cancer cell ,Cancer research - Abstract
Purpose: hERG1 channels are aberrantly expressed in several types of human cancers, where they affect different aspects of cancer cell behavior. A thorough analysis of the functional role and clinical significance of hERG1 channels in gastric cancer is still lacking. Experimental Design: hERG1 expression was tested in a wide (508 samples) Italian cohort of surgically resected patients with gastric cancer, by immunohistochemistry and real-time quantitative PCR. The functional link between hERG1 and the VEGF-A was studied in different gastric cancer cell lines. The effects of hERG1 and VEGF-A inhibition were evaluated in vivo in xenograft mouse models. Results: hERG1 was positive in 69% of the patients and positivity correlated with Lauren's intestinal type, fundus localization of the tumor, G1–G2 grading, I and II tumor—node—metastasis stage, and VEGF-A expression. hERG1 activity modulated VEGF-A secretion, through an AKT-dependent regulation of the transcriptional activity of the hypoxia inducible factor. Treatment of immunodeficient mice xenografted with human gastric cancer cells, with a combination of hERG1 blockers and anti-VEGF-A antibodies, impaired tumor growth more than single-drug treatments. Conclusion: Our results show that hERG1 (i) is aberrantly expressed in human gastric cancer since its early stages; (ii) drives an intracellular pathway leading to VEGF-A secretion; (iii) can be exploited to identify a gastric cancer patients' group where a combined treatment with antiangiogenic drugs and noncardiotoxic hERG1 inhibitors could be proposed. Clin Cancer Res; 20(6); 1502–12. ©2014 AACR.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.