48 results on '"Gelsomino L"'
Search Results
2. Farnesoid X receptor inhibits tamoxifen-resistant MCF-7 breast cancer cell growth through downregulation of HER2 expression
- Author
-
Giordano, C, Catalano, S, Panza, S, Vizza, D, Barone, I, Bonofiglio, D, Gelsomino, L, Rizza, P, Fuqua, S A W, and Andò, S
- Published
- 2011
- Full Text
- View/download PDF
3. Abstract P1-05-04: Leptin modulates exosome biogenesis in breast cancer cells through an enhanced Hsp90/Tsg101 interaction
- Author
-
Gelsomino, L, primary, Giordano, C, additional, Barone, I, additional, Panza, S, additional, Augimeri, G, additional, Bonofiglio, D, additional, Catalano, S, additional, and Andò, S, additional
- Published
- 2019
- Full Text
- View/download PDF
4. Abstract S4-02: The Y537S ESR1 mutation is a dominant driver of distant ER-positive breast cancer metastasis
- Author
-
Fuqua, SAW, primary, Gu, G, additional, Rechoum, Y, additional, Gelsomino, L, additional, Dustin, DJ, additional, Corona-Rodriguez, A, additional, Beyer, AR, additional, Pejerrey, SM, additional, Gao, M, additional, Tsimelzon, A, additional, Tian, L, additional, Zhang, X, additional, Nagi, C, additional, and Ando', S, additional
- Published
- 2017
- Full Text
- View/download PDF
5. Introducing a financial perspective in Supply Chain Management: a literature review on Supply Chain Finance
- Author
-
Gelsomino, L. M., Mangiaracina, R., Perego, A., and ANGELA TUMINO
- Published
- 2014
6. Structural, Thermodynamic, and Kinetic Traits of Antiestrogen-Compounds Selectively Targeting the Y537S Mutant Estrogen Receptor α Transcriptional Activity in Breast Cancer Cell Lines
- Author
-
Alessandra Magistrato, Stefania Catalano, Angelo Spinello, Luca Gelsomino, Ines Barone, Sebastiano Andò, Matic Pavlin, Pavlin M., Gelsomino L., Barone I., Spinello A., Catalano S., Ando S., and Magistrato A.
- Subjects
medicine.drug_class ,Somatic cell ,In silico ,Estrogen receptor ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,lcsh:Chemistry ,Breast cancer ,breast cancer ,medicine ,Aromatase ,resistant breast cancers ,Original Research ,biology ,Chemistry ,Wild type ,Y537S ,General Chemistry ,021001 nanoscience & nanotechnology ,Antiestrogen ,medicine.disease ,SERM ,molecular dynamics ,0104 chemical sciences ,lcsh:QD1-999 ,Estrogen ,SERD ,biology.protein ,Cancer research ,0210 nano-technology ,estrogen receptor - Abstract
The most frequently diagnosed cancers in women are the estrogen receptor (ER)-positive breast cancer subtypes, which are characterized by estrogen dependency for their growth. The mainstay of clinical treatment for this tumor relies on the modulation of ER action or on the suppression of estrogen biosynthesis via the administration of Selective ERα Modulators/Down-regulators (SERMs/SERDs) or aromatase inhibitors, respectively. Nevertheless, de novo and acquired resistance to these therapies frequently occurs and represents a major clinical concern for patient survival. Recently, somatic mutations affecting the hormone-binding domain of ERα (i. e. Y537S, Y537N, D538G) have been associated with endocrine resistance, disease relapse and increased mortality rates. Hence, devising novel therapies against these ERα isoforms represents a daunting challenge. Here, we identified five molecules active on recurrent Y537S ERα polymorphism by employing in silico virtual screening on commercial databases of molecules, complemented by ER-transactivation and MTT assays in MCF7 and MDA-MB-231 breast cancer cells expressing wild type or mutated ERα. Among them, one molecule selectively targets Y537S ERα without inducing any cytotoxicity in breast cell lines. Multi-microseconds (4.5 µs) of biased and unbiased molecular dynamics provided an atomic-level picture of the structural, thermodynamics (i. e. binding free energies) and the kinetic (i. e. dissociation free energy barriers) of these active ligands as compared to clinically used SERM/SERDs upon binding to wild type and distinct ERα variants (Y537S, Y537N, D538G). This study contributes to a dissection of the key molecular traits needed by drug-candidates to hamper the agonist (active)-like conformation of ERα, normally selected by those polymorphic variants. This information can be useful to discover mutant specific drug-candidates, enabling to move a step forward towards tailored approaches for breast cancer treatment.
- Published
- 2019
- Full Text
- View/download PDF
7. Verso un Parco culturale della Linea Gotica in Toscana
- Author
-
GELSOMINO, LUISELLA and Gelsomino L., (a cura di)
- Subjects
LINEA GOTICA ,PAESAGGIO ,BENI CULTURALI ,PARCO CULTURALE ,RICERCA STORICA - Published
- 2006
8. Le Reggiane - Area strategica tra vecchia e nuova identità urbana
- Author
-
GELSOMINO, LUISELLA and Gelsomino L., (a cura di)
- Subjects
RINNOVO URBANO ,PAESAGGIO ,BENI CULTURALI ,PIANO TERRITORIALE PAESISTICO ,QUALITÀ URBANA - Abstract
Sulla base di quanto previsto all’art. 8 dell’Accordo siglato il 19 aprile 2001 dal Ministro per i Beni e le Attività Culturali e dalle Regioni e in riferimento alle disposizioni contenute nel Codice dei beni culturali e del paesaggio approvato dal Consiglio dei Ministri il 22 gennaio 2004, lo studio intende fornire elementi di riflessione e proposte concrete per la formulazione di indicazioni relative all’adeguamento del Piano territoriale paesistico della Regione Emilia-Romagna (Ptpr).
- Published
- 2006
9. Paesaggi della memoria - Itinerari della Linea Gotica in Toscana
- Author
-
GELSOMINO, LUISELLA, Altri, Gelsomino L., and Altri
- Subjects
LINEA GOTICA ,PATRIMONIO STORICO ,PAESAGGIO ,PARCO CULTURALE ,ITINERARI - Abstract
Memorie, tracce, presenze della Linea Gotica in Toscana sono scrupolosamente ricostruite in questa guida che provincia per provincia - da Massa a Carrara, da Lucca a Pistoia, a Prato, a Firenze, ad Arezzo - affianca due strade: la rigorosa ricostruzione storica delle vicende di guerrra e la proposta di itinerari tematici di visita, indispensabili per l'individuazione e la concreta fruizione turistica dei luoghi. Riferimenti informativi dettagliati ed esaurienti accompagnano questo viaggio nella Memoria.
- Published
- 2005
10. Evidence that CRISPR-Cas9 Y537S-mutant expressing breast cancer cells activate Yes-associated protein 1 to driving the conversion of normal fibroblasts into cancer-associated fibroblasts.
- Author
-
Gelsomino L, Caruso A, Tasan E, Leonetti AE, Malivindi R, Naimo GD, Giordano F, Panza S, Gu G, Perrone B, Giordano C, Mauro L, Nardo B, Filippelli G, Bonofiglio D, Barone I, Fuqua SAW, Catalano S, and Andò S
- Subjects
- Humans, Animals, Female, Mice, Mice, Nude, MCF-7 Cells, Fibroblasts metabolism, Cell Proliferation, Transcription Factors metabolism, Transcription Factors genetics, Tumor Microenvironment genetics, Estrogen Receptor alpha metabolism, Estrogen Receptor alpha genetics, Cell Movement genetics, Receptor, IGF Type 1 metabolism, Receptor, IGF Type 1 genetics, Breast Neoplasms pathology, Breast Neoplasms genetics, Breast Neoplasms metabolism, YAP-Signaling Proteins, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, CRISPR-Cas Systems genetics, Mutation genetics, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics
- Abstract
Background: Endocrine therapy (ET) has improved the clinical outcomes of Estrogen receptor alpha-positive (ERɑ +) breast cancer (BC) patients, even though resistance to ET remains a clinical issue. Mutations in the hormone-binding domain of ERɑ represent an acquired intrinsic mechanism of ET resistance. However, the latter also depends on the multiple functional interactions between BC cells and the tumor microenvironment (TME). Here, we investigated how the most common Y537S-ERɑ mutation may influence the behavior of fibroblasts, the most prominent component of the TME., Methods: We conducted coculture experiments with normal human foreskin fibroblasts BJ1-hTERT (NFs), cancer-associated fibroblasts (CAFs), isolated from human BC specimens, and Y537S CRISPR-expressing MCF-7 BC cells (MCF-7YS). Mass spectrometry (MS) and Metacore analyses were performed to investigate how the functional interactions between BC cells/fibroblasts may affect their proteomic profile. The impact of fibroblasts on BC tumor growth and metastatic potential was evaluated in nude mice., Results: Mutant BC conditioned medium (CM) affected the morphology/proliferation/migration of both NFs and CAFs. 198 deregulated proteins signed the proteomic similarity profile of NFs exposed to the YS-CM and CAFs. Among the upregulated proteins, Yes-associated protein 1 (YAP1) was the main central hub in the direct interaction network. Increased YAP1 protein expression and activity were confirmed in NFs treated with MCF-7YS-CM. However, YAP1 activation appears to crosstalk with the insulin growth factor-1 receptor (IGF-1R). Higher amount of IGF-1 were noticed in the MCF-7YS-CM cells compared to the MCF-7P, and IGF-1 immunodepletion reversed the enhanced YAP1 expression and activity. Mutant cells upon exposure to the NF- and CAF-CM exhibited an enhanced proliferation/growth/migration/invasion compared to the MCF-7P. MCF-7YS cells when implanted with CAFs showed an early relative increased tumor volume compared to YS alone. No changes were observed when MCF-7P cells were co-implanted with CAFs. Compared with that in MCF-7P cells, the metastatic burden of MCF-7YS cells was intrinsically greater, and this effect was augmented upon treatment with NF-CM and further increased with CAF-CM., Conclusions: YS mutant BC cells induced the conversion of fibroblasts into CAFs, via YAP, which represent a potential therapeutic target which interrupt the functional interactions between mutant cells/TME and to be implemented in the novel therapeutic strategy of a subset of metastatic BC patients carrying the frequent Y537S mutations., Competing Interests: Declarations Ethics approval and consent to participate In the present study we used fibroblast specimens, isolated in accordance with approved guidelines, from patients who had signed informed consent and approved by the Ethic Institutional Committees at Annunziata Hospital, Cosenza, Italy (#149 issued by Comitato Etico Regione Calabria, Sezione Area Nord c/o Azienda Ospedaliera di Cosenza, 28/10/2015). All animals were maintained and handled in accordance with the recommendation of the Guidelines for the Care and Use of Laboratory Animals and experiments were approved by the Animal Care Committee of University of Calabria (OPBA), Italy (454/2023-PR, May 17, 2023). Consent for publication We have obtained consents to publish this paper from all the participants of this study. Competing interests The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. miRNAs in the Box: Potential Diagnostic Role for Extracellular Vesicle-Packaged miRNA-27a and miRNA-128 in Breast Cancer.
- Author
-
Giordano C, Accattatis FM, Gelsomino L, Del Console P, Győrffy B, Giuliano M, Veneziani BM, Arpino G, De Angelis C, De Placido P, Pietroluongo E, Zinno F, Bonofiglio D, Andò S, Barone I, and Catalano S
- Subjects
- Humans, Female, MicroRNAs metabolism, Breast Neoplasms diagnosis, Breast Neoplasms genetics, Breast Neoplasms metabolism, Extracellular Vesicles genetics, Extracellular Vesicles metabolism
- Abstract
Circulating extracellular vesicle (EV)-derived microRNAs (miRNAs) are now considered the next generation of cancer "theranostic" tools, with strong clinical relevance. Although their potential in breast cancer diagnosis has been widely reported, further studies are still required to address this challenging issue. The present study examined the expression profiles of EV-packaged miRNAs to identify novel miRNA signatures in breast cancer and verified their diagnostic accuracy. Circulating EVs were isolated from healthy controls and breast cancer patients and characterized following the MISEV 2018 guidelines. RNA-sequencing and real-time PCR showed that miRNA-27a and miRNA-128 were significantly down-regulated in patient-derived EVs compared to controls in screening and validation cohorts. Bioinformatics analyses of miRNA-target genes indicated several enriched biological processes/pathways related to breast cancer. Receiver operating characteristic (ROC) curves highlighted the ability of these EV-miRNAs to distinguish breast cancer patients from non-cancer controls. According to other reports, the levels of EV-miRNA-27a and EV-miRNA-128 are not associated with their circulating ones. Finally, evidence from the studies included in our systematic review underscores how the expression of these miRNAs in biofluids is still underinvestigated. Our findings unraveled the role of serum EV-derived miRNA-27a and miRNA-128 in breast cancer, encouraging further investigation of these two miRNAs within EVs towards improved breast cancer detection.
- Published
- 2023
- Full Text
- View/download PDF
12. ERα/LKB1 complex upregulates E-cadherin expression and stimulates breast cancer growth and progression upon adiponectin exposure.
- Author
-
Naimo GD, Forestiero M, Paolì A, Malivindi R, Gelsomino L, Győrffy B, Leonetti AE, Giordano F, Panza S, Conforti FL, Ruffo P, Panno ML, Mauro L, and Andò S
- Subjects
- Humans, Animals, Mice, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Cell Line, Tumor, MCF-7 Cells, Cadherins genetics, Adiponectin, Neoplasms
- Abstract
Adiponectin is the major adipocytes-secreted protein involved in obesity-related breast cancer growth and progression. We proved that adiponectin promotes proliferation in ERα-positive breast cancer cells, through ERα transactivation and the recruitment of LKB1 as ERα-coactivator. Here, we showed that adiponectin-mediated ERα transactivation enhances E-cadherin expression. Thus, we investigated the molecular mechanism through which ERα/LKB1 complex may modulate the expression of E-cadherin, influencing tumor growth, progression and distant metastasis. We demonstrated that adiponectin increases E-cadherin expression in ERα-positive 2D and higher extent in 3D cultures. This occurs through a direct activation of E-cadherin gene promoter by ERα/LKB1-complex. The impact of E-cadherin on ERα-positive breast cancer cell proliferation comes from the evidence that in the presence of E-cadherin siRNA the proliferative effects of adiponectin is no longer noticeable. Since E-cadherin connects cell polarity and growth, we investigated if the adiponectin-enhanced E-cadherin expression could influence the localization of proteins cooperating in cell polarity, such as LKB1 and Cdc42. Surprisingly, immunofluorescence showed that, in adiponectin-treated MCF-7 cells, LKB1 and Cdc42 mostly colocalize in the nucleus, impairing their cytosolic cooperation in maintaining cell polarity. The orthotopic implantation of MCF-7 cells revealed an enhanced E-cadherin-mediated breast cancer growth induced by adiponectin. Moreover, tail vein injection of MCF-7 cells showed a higher metastatic burden in the lungs of mice receiving adiponectin-treated cells compared to control. From these findings it emerges that adiponectin treatment enhances E-cadherin expression, alters cell polarity and stimulates ERα-positive breast cancer cell growth in vitro and in vivo, sustaining higher distant metastatic burden., (© 2023 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)
- Published
- 2023
- Full Text
- View/download PDF
13. Leptin: A Heavyweight Player in Obesity-Related Cancers.
- Author
-
Caruso A, Gelsomino L, Panza S, Accattatis FM, Naimo GD, Barone I, Giordano C, Catalano S, and Andò S
- Subjects
- Humans, Adipose Tissue metabolism, Obesity complications, Obesity metabolism, Adipokines metabolism, Leptin metabolism, Neoplasms metabolism
- Abstract
Obesity, defined as the abnormal or excessive expansion of white adipose tissue, has reached pandemic proportions and is recognized as an important health concern since it is a common root for several comorbidities, including malignancies. Indeed, the current knowledge of the white adipose tissue, which shifts its role from an energy storage tissue to an important endocrine and metabolic organ, has opened up new avenues for the discovery of obesity's effects on tumor biology. In this review, we will report the epidemiological studies concerning the strong impact of obesity in several types of cancer and describe the mechanisms underlying the heterotypic signals between cancer cell lines and adipocytes, with particular emphasis on inflammation, the insulin/IGF-1 axis, and adipokines. Among the adipokines, we will further describe the in vitro, in vivo, and clinical data concerning the role of leptin, recognized as one of the most important mediators of obesity-associated cancers. In fact, leptin physiologically regulates energy metabolism, appetite, and reproduction, and several studies have also described the role of leptin in affecting cancer development and progression. Finally, we will summarize the newest pharmacological strategies aimed at mitigating the protumorigenic effects of leptin, underlining their mechanisms of action.
- Published
- 2023
- Full Text
- View/download PDF
14. CEBP-β and PLK1 as Potential Mediators of the Breast Cancer/Obesity Crosstalk: In Vitro and In Silico Analyses.
- Author
-
Accattatis FM, Caruso A, Carleo A, Del Console P, Gelsomino L, Bonofiglio D, Giordano C, Barone I, Andò S, Bianchi L, and Catalano S
- Subjects
- Female, Humans, MCF-7 Cells, Adipocytes metabolism, Adipose Tissue metabolism, Obesity metabolism, Cell Proliferation, Cell Line, Tumor, Breast Neoplasms metabolism
- Abstract
Over the last two decades, obesity has reached pandemic proportions in several countries, and expanding evidence is showing its contribution to several types of malignancies, including breast cancer (BC). The conditioned medium (CM) from mature adipocytes contains a complex of secretes that may mimic the obesity condition in studies on BC cell lines conducted in vitro. Here, we report a transcriptomic analysis on MCF-7 BC cells exposed to adipocyte-derived CM and focus on the predictive functional relevance that CM-affected pathways/processes and related biomarkers (BMs) may have in BC response to obesity. CM was demonstrated to increase cell proliferation, motility and invasion as well as broadly alter the transcript profiles of MCF-7 cells by significantly modulating 364 genes. Bioinformatic functional analyses unraveled the presence of five highly relevant central hubs in the direct interaction networks (DIN), and Kaplan-Meier analysis sorted the CCAAT/enhancer binding protein beta (CEBP-β) and serine/threonine-protein kinase PLK1 (PLK1) as clinically significant biomarkers in BC. Indeed, CEBP-β and PLK1 negatively correlated with BC overall survival and were up-regulated by adipocyte-derived CM. In addition to their known involvement in cell proliferation and tumor progression, our work suggests them as a possible "deus ex machina" in BC response to fat tissue humoral products in obese women.
- Published
- 2023
- Full Text
- View/download PDF
15. Analysis of circulating extracellular vesicle derived microRNAs in breast cancer patients with obesity: a potential role for Let-7a.
- Author
-
Barone I, Gelsomino L, Accattatis FM, Giordano F, Gyorffy B, Panza S, Giuliano M, Veneziani BM, Arpino G, De Angelis C, De Placido P, Bonofiglio D, Andò S, Giordano C, and Catalano S
- Subjects
- Humans, Female, Phosphatidylinositol 3-Kinases metabolism, Quality of Life, Obesity complications, Obesity genetics, Obesity metabolism, Circulating MicroRNA metabolism, Breast Neoplasms complications, Breast Neoplasms genetics, Breast Neoplasms metabolism, MicroRNAs metabolism, Extracellular Vesicles metabolism
- Abstract
Background: The incidence of obesity, a known risk factor for several metabolic and chronic diseases, including numerous malignancies, has risen sharply in the world. Various clinical studies demonstrate that excessive Body Mass Index (BMI) may worsen the incidence, prognosis, and mortality rates of breast cancer. Thus, understanding the link tying up obesity and breast cancer onset and progression is critically important, as it can impact patients' survival and quality of life. Recently, circulating extracellular vesicle (EV) derived miRNAs have attracted much attention for their diagnostic, prognostic and therapeutic potential in oncology research. Although the potential role of EV-derived miRNAs in the early detection of breast cancer has been repeatedly mentioned, screening of miRNAs packaged within serum EVs has not yet been reported in patients with obesity., Methods: Circulating EVs were isolated from normal weight (NW), and overweight/obese (OW/Ob) breast cancer patients and characterized by Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and protein marker expression. Evaluation of EV-associated miRNAs was conducted in a screening (RNA-seq) and a validation (qRT-PCR) cohort. Bioinformatic analysis was performed to uncover significantly enriched biological processes, molecular functions and pathways. ROC and Kaplain-Meier survival analyses were used for clinical significance., Results: Comparison of serum EV-derived miRNAs from NW and OW/Ob patients detected seven differentially expressed miRNAs (let-7a-5p, miR-122-5p, miR-30d-5p, miR-126-3p, miR-27b-3p, miR-4772-3p, and miR-10a-5p) in the screening cohort. GO analysis revealed the enrichment of protein phosphorylation, intracellular signal transduction, signal transduction, and vesicle-mediated transport among the top biological processes. In addition, the target genes were significantly enriched in pathways related to PI3K/Akt, growth hormones, and insulin signalings, which are all involved in obesity-related diseases and/or breast cancer progression. In the validation cohort, qRT-PCR confirmed a significant down-regulation of EV-derived let-7a in the serum of OW/Ob breast cancer patients compared to NW patients. Let-7a levels also exhibited a negative correlation with BMI values. Importantly, decreased let-7a miRNA expression was associated with higher tumor grade and poor survival in patients with breast cancer., Conclusion: These results suggest that serum-EV derived miRNAs may reflect a differential profile in relation to a patient's BMI, which, once validated in larger cohorts of patients, could provide insights into novel specific biomarkers and innovative targets to prevent the progression of obesity-mediated breast cancer., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
16. Proteomic Profiling of Extracellular Vesicles Released by Leptin-Treated Breast Cancer Cells: A Potential Role in Cancer Metabolism.
- Author
-
Gelsomino L, Barone I, Caruso A, Giordano F, Brindisi M, Morello G, Accattatis FM, Panza S, Cappello AR, Bonofiglio D, Andò S, Catalano S, and Giordano C
- Subjects
- Humans, Female, Proteomics, Leptin metabolism, Chromatography, Liquid, Tandem Mass Spectrometry, Obesity metabolism, Tumor Microenvironment, Breast Neoplasms metabolism, Extracellular Vesicles metabolism
- Abstract
Tumor extracellular vesicles (EVs), as endocytic vesicles able to transport nucleic acids, proteins, and metabolites in recipient cells, have been recognized fundamental mediators of cell-to-cell communication in breast cancer. The biogenesis and release of EVs are highly regulated processes and both the quantity of EVs and their molecular cargo might reflect the metabolic state of the producing cells. We recently demonstrated that the adipokine leptin, whose circulating levels correlate with adipose tissue expansion, is an inducer of EV release from breast cancer cells. Here, we show a specific proteomic signature of EVs released by MCF-7 breast cancer cells grown in the presence of leptin (Lep-EVs), in attempt to find additional molecular effectors linking obesity to breast cancer biology. An analysis of the proteomic profile of Lep-EVs by LC-MS/MS revealed a significant enrichment in biological processes, molecular functions, and cellular components mainly related to mitochondrial machineries and activity, compared to protein content of EVs from untreated breast cancer cells. Metabolic investigations, carried out to assess the autocrine effects of these vesicles on breast cancer cells, revealed that Lep-EVs were able to increase ATP levels in breast cancer cells. This result is associated with increased mitochondrial respiration evaluated by Seahorse analyzer, supporting the concept that Lep-EVs can modulate MCF-7 breast cancer cell oxidative metabolism. Moreover, taking into account the relevance of tumor immune cell crosstalk in the tumor microenvironment (TME), we analyzed the impact of these vesicles on macrophage polarization, the most abundant immune component in the breast TME. We found that tumor-derived Lep-EVs sustain the polarization of M0 macrophages, derived from the human THP-1 monocytic cells, into M2-like tumor-associated macrophages, in terms of metabolic features, phagocytic activity, and increased expression of CD206-positive population. Overall, our results indicate that leptin by inducing the release of EV-enriched in mitochondrial proteins may control the metabolism of MCF-7 breast cancer cells as well as that of macrophages. Characterization of tumor-derived EV protein cargo in an obesity-associated milieu, such as in the presence of elevated leptin levels, might allow identifying unique features and specific metabolic mechanisms useful to develop novel therapeutic approaches for treatment of breast cancer, especially in obese patients.
- Published
- 2022
- Full Text
- View/download PDF
17. Obesity and endocrine therapy resistance in breast cancer: Mechanistic insights and perspectives.
- Author
-
Barone I, Caruso A, Gelsomino L, Giordano C, Bonofiglio D, Catalano S, and Andò S
- Subjects
- Aromatase Inhibitors adverse effects, Female, Humans, Obesity complications, Obesity drug therapy, Receptors, Estrogen, Selective Estrogen Receptor Modulators adverse effects, Breast Neoplasms drug therapy, Breast Neoplasms pathology
- Abstract
The incidence of obesity, a recognized risk factor for various metabolic and chronic diseases, including numerous types of cancers, has risen dramatically over the recent decades worldwide. To date, convincing research in this area has painted a complex picture about the adverse impact of high body adiposity on breast cancer onset and progression. However, an emerging but overlooked issue of clinical significance is the limited efficacy of the conventional endocrine therapies with selective estrogen receptor modulators (SERMs) or degraders (SERDs) and aromatase inhibitors (AIs) in patients affected by breast cancer and obesity. The mechanisms behind the interplay between obesity and endocrine therapy resistance are likely to be multifactorial. Therefore, what have we actually learned during these years and which are the main challenges in the field? In this review, we will critically discuss the epidemiological evidence linking obesity to endocrine therapeutic responses and we will outline the molecular players involved in this harmful connection. Given the escalating global epidemic of obesity, advances in understanding this critical node will offer new precision medicine-based therapeutic interventions and more appropriate dosing schedule for treating patients affected by obesity and with breast tumors resistant to endocrine therapies., (© 2021 The Authors. Obesity Reviews published by John Wiley & Sons Ltd on behalf of World Obesity Federation.)
- Published
- 2022
- Full Text
- View/download PDF
18. Correction to: Hormonal modulation of ESR1 mutant metastasis.
- Author
-
Gu G, Tian L, Herzog SK, Rechoum Y, Gelsomino L, Gao M, Du L, Kim JA, Dustin D, Lo HC, Beyer AR, Edwards DG, Gonzalez T, Tsimelzon A, Huang HJ, Fernandez NM, Grimm SL, Hilsenbeck SG, Liu D, Xu J, Alaniz A, Li S, Mills GB, Janku F, Kittler R, Zhang XH, Coarfa C, Foulds CE, Symmans WF, Andò S, and Fuqua SAW
- Published
- 2022
- Full Text
- View/download PDF
19. Novel Insights into the Antagonistic Effects of Losartan against Angiotensin II/AGTR1 Signaling in Glioblastoma Cells.
- Author
-
Panza S, Malivindi R, Caruso A, Russo U, Giordano F, Győrffy B, Gelsomino L, De Amicis F, Barone I, Conforti FL, Giordano C, Bonofiglio D, Catalano S, and Andò S
- Abstract
New avenues for glioblastoma therapy are required due to the limited mortality benefit of the current treatments. The renin-angiotensin system (RAS) exhibits local actions and works as a paracrine system in different tissues and tumors, including glioma. The glioblastoma cell lines U-87 MG and T98G overexpresses Angiotensin II (Ang II)/Angiotensin II type I receptor (AGTR1) signaling, which enhances in vitro and in vivo local estrogen production through a direct up-regulation of the aromatase gene promoters p I.f and p I.4. In addition, Ang II/AGTR1 signaling transactivates estrogen receptor-α in a ligand-independent manner through mitogen-activated protein kinase (MAPK) activation. The higher aromatase mRNA expression in patients with glioblastoma was associated with the worst survival prognostic, according to The Cancer Genome Atlas (TCGA). An intrinsic immunosuppressive glioblastoma tumor milieu has been previously documented. We demonstrate how Ang II treatment in glioblastoma cells increases programmed death-ligand 1 (PD-L1) expression reversed by combined exposure to Losartan (LOS) in vitro and in vivo. Our findings highlight how LOS, in addition, antagonizes the previously documented neoangiogenetic, profibrotic, and immunosuppressive effects of Ang II and drastically inhibits its stimulatory effects on local estrogen production, sustaining glioblastoma cell growth. Thus, Losartan may represent an adjuvant pharmacological tool to be repurposed prospectively for glioblastoma treatment.
- Published
- 2021
- Full Text
- View/download PDF
20. Adipocyte-derived extracellular vesicles promote breast cancer cell malignancy through HIF-1α activity.
- Author
-
La Camera G, Gelsomino L, Malivindi R, Barone I, Panza S, De Rose D, Giordano F, D'Esposito V, Formisano P, Bonofiglio D, Andò S, Giordano C, and Catalano S
- Abstract
Extracellular vesicles (EVs) are emerging key protagonists in intercellular communication between adipocytes and breast cancer (BC) cells. Here, we described a new mechanism by which EVs released by mature adipocytes promoted breast cancer cell malignancy "in vitro" and "in vivo". We found that adipocyte-derived EVs enhanced growth, motility and invasion, stem cell-like properties, as well as specific traits of epithelial-to-mesenchymal transition in both estrogen receptor positive and triple negative BC cells. Of note, adipocyte-derived EVs aid breast tumor cells in lung metastatic colonization after tail-vein injection in mice. These EV-mediated effects occur via the induction of HIF-1α activity, since they were abrogated by the use of the HIF-1α inhibitor KC7F2 or in cells silenced for HIF-1α expression. Moreover, using an "ex vivo" model of obese adipocytes we found that the depletion of EVs counteracted the ability of obese adipocytes to sustain pro-invasive phenotype in BC cells. Interestingly, EVs released by undifferentiated adipocytes failed to induce aggressiveness and HIF-1α expression. These findings shed new light on the role of adipocyte-derived EVs in breast cancer progression, suggesting the possibility to target HIF-1α activity to block the harmful adipocyte-tumor cell dialogue, especially in obese settings., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. The Emerging Role of Extracellular Vesicles in Endocrine Resistant Breast Cancer.
- Author
-
La Camera G, Gelsomino L, Caruso A, Panza S, Barone I, Bonofiglio D, Andò S, Giordano C, and Catalano S
- Abstract
Breast cancer is the most common solid malignancy diagnosed in females worldwide, and approximately 70% of these tumors express estrogen receptor α (ERα), the main biomarker of endocrine therapy. Unfortunately, despite the use of long-term anti-hormone adjuvant treatment, which has significantly reduced patient mortality, resistance to the endocrine treatments often develops, leading to disease recurrence and limiting clinical benefits. Emerging evidence indicates that extracellular vesicles (EVs), nanosized particles that are released by all cell types and responsible for local and systemic intercellular communications, might represent a newly identified mechanism underlying endocrine resistance. Unraveling the role of EVs, released by transformed cells during the tumor evolution under endocrine therapy, is still an open question in the cancer research area and the molecular mechanisms involved should be better defined to discover alternative therapeutic approaches to overcome resistance. In this review, we will provide an overview of recent findings on the involvement of EVs in sustaining hormonal resistance in breast cancer and discuss opportunities for their potential use as biomarkers to monitor the therapeutic response and disease progression.
- Published
- 2021
- Full Text
- View/download PDF
22. Hormonal modulation of ESR1 mutant metastasis.
- Author
-
Gu G, Tian L, Herzog SK, Rechoum Y, Gelsomino L, Gao M, Du L, Kim JA, Dustin D, Lo HC, Beyer AR, Edwards DG, Gonzalez T, Tsimelzon A, Huang HJ, Fernandez NM, Grimm SL, Hilsenbeck SG, Liu D, Xu J, Alaniz A, Li S, Mills GB, Janku F, Kittler R, Zhang XH, Coarfa C, Foulds CE, Symmans WF, Andò S, and Fuqua SAW
- Subjects
- Animals, Aromatase Inhibitors pharmacology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Dihydrotestosterone pharmacology, Estradiol metabolism, Estrogens genetics, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Mutation genetics, Neoplasm Metastasis, Receptors, Androgen drug effects, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Estrogen Receptor alpha genetics, Receptors, Androgen genetics, Tamoxifen pharmacology
- Abstract
Estrogen receptor alpha gene (ESR1) mutations occur frequently in ER-positive metastatic breast cancer, and confer clinical resistance to aromatase inhibitors. Expression of the ESR1 Y537S mutation induced an epithelial-mesenchymal transition (EMT) with cells exhibiting enhanced migration and invasion potential in vitro. When small subpopulations of Y537S ESR1 mutant cells were injected along with WT parental cells, tumor growth was enhanced with mutant cells becoming the predominant population in distant metastases. Y537S mutant primary xenograft tumors were resistant to the antiestrogen tamoxifen (Tam) as well as to estradiol (E
2 ) withdrawal. Y537S ESR1 mutant primary tumors metastasized efficiently in the absence of E2 ; however, Tam treatment significantly inhibited metastasis to distant sites. We identified a nine-gene expression signature, which predicted clinical outcomes of ER-positive breast cancer patients, as well as breast cancer metastasis to the lung. Androgen receptor (AR) protein levels were increased in mutant models, and the AR agonist dihydrotestosterone significantly inhibited estrogen-regulated gene expression, EMT, and distant metastasis in vivo, suggesting that AR may play a role in distant metastatic progression of ESR1 mutant tumors.- Published
- 2021
- Full Text
- View/download PDF
23. Natural and Synthetic PPARγ Ligands in Tumor Microenvironment: A New Potential Strategy against Breast Cancer.
- Author
-
Augimeri G, Gelsomino L, Plastina P, Giordano C, Barone I, Catalano S, Andò S, and Bonofiglio D
- Subjects
- Animals, Breast Neoplasms immunology, Breast Neoplasms metabolism, Female, Humans, Ligands, Signal Transduction, Breast Neoplasms pathology, PPAR gamma metabolism, Tumor Microenvironment immunology
- Abstract
Multiple lines of evidence indicate that activation of the peroxisome proliferator-activated receptor γ (PPARγ) by natural or synthetic ligands exerts tumor suppressive effects in different types of cancer, including breast carcinoma. Over the past decades a new picture of breast cancer as a complex disease consisting of neoplastic epithelial cells and surrounding stroma named the tumor microenvironment (TME) has emerged. Indeed, TME is now recognized as a pivotal element for breast cancer development and progression. Novel strategies targeting both epithelial and stromal components are under development or undergoing clinical trials. In this context, the aim of the present review is to summarize PPARγ activity in breast TME focusing on the role of this receptor on both epithelial/stromal cells and extracellular matrix components of the breast cancer microenvironment. The information provided from the in vitro and in vivo research indicates PPARγ ligands as potential agents with regards to the battle against breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
24. The Role of PPARγ Ligands in Breast Cancer: From Basic Research to Clinical Studies.
- Author
-
Augimeri G, Giordano C, Gelsomino L, Plastina P, Barone I, Catalano S, Andò S, and Bonofiglio D
- Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), belonging to the nuclear receptor superfamily, is a ligand-dependent transcription factor involved in a variety of pathophysiological conditions such as inflammation, metabolic disorders, cardiovascular disease, and cancers. In this latter context, PPARγ is expressed in many tumors including breast cancer, and its function upon binding of ligands has been linked to the tumor development, progression, and metastasis. Over the last decade, much research has focused on the potential of natural agonists for PPARγ including fatty acids and prostanoids that act as weak ligands compared to the strong and synthetic PPARγ agonists such as thiazolidinedione drugs. Both natural and synthetic compounds have been implicated in the negative regulation of breast cancer growth and progression. The aim of the present review is to summarize the role of PPARγ activation in breast cancer focusing on the underlying cellular and molecular mechanisms involved in the regulation of cell proliferation, cell cycle, and cell death, in the modulation of motility and invasion as well as in the cross-talk with other different signaling pathways. Besides, we also provide an overview of the in vivo breast cancer models and clinical studies. The therapeutic effects of natural and synthetic PPARγ ligands, as antineoplastic agents, represent a fascinating and clinically a potential translatable area of research with regards to the battle against cancer.
- Published
- 2020
- Full Text
- View/download PDF
25. Evidence for Enhanced Exosome Production in Aromatase Inhibitor-Resistant Breast Cancer Cells.
- Author
-
Augimeri G, La Camera G, Gelsomino L, Giordano C, Panza S, Sisci D, Morelli C, Győrffy B, Bonofiglio D, Andò S, Barone I, and Catalano S
- Subjects
- Aromatase Inhibitors pharmacology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cluster Analysis, Estrogens deficiency, Exosomes ultrastructure, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Proteomics, Up-Regulation drug effects, rab GTP-Binding Proteins metabolism, Aromatase Inhibitors therapeutic use, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm drug effects, Exosomes metabolism
- Abstract
Aromatase inhibitors (AIs) represent the standard anti-hormonal therapy for post-menopausal estrogen receptor-positive breast cancer, but their efficacy is limited by the emergence of AI resistance (AI
R ). Exosomes act as vehicles to engender cancer progression and drug resistance. The goal of this work was to study exosome contribution in AIR mechanisms, using estrogen-dependent MCF-7 breast cancer cells as models and MCF-7 LTED (Long-Term Estrogen Deprived) subline, modeling AIR . We found that exosome secretion was significantly increased in MCF-7 LTED cells compared to MCF-7 cells. MCF-7 LTED cells also exhibited a higher amount of exosomal RNA and proteins than MCF-7 cells. Proteomic analysis revealed significant alterations in the cellular proteome. Indeed, we showed an enrichment of proteins frequently identified in exosomes in MCF-7 LTED cells. The most up-regulated proteins in MCF-7 LTED cells were represented by Rab GTPases, important vesicle transport-regulators in cancer, that are significantly mapped in "small GTPase-mediated signal transduction", "protein transport" and "vesicle-mediated transport" Gene Ontology categories. Expression of selected Rab GTPases was validated by immunoblotting. Collectively, we evidence, for the first time, that AIR breast cancer cells display an increased capability to release exosomes, which may be associated with an enhanced Rab GTPase expression. These data provide the rationale for further studies directed at clarifying exosome's role on endocrine therapy, with the aim to offer relevant markers and druggable therapeutic targets for the management of hormone-resistant breast cancers.- Published
- 2020
- Full Text
- View/download PDF
26. The Biology of Exosomes in Breast Cancer Progression: Dissemination, Immune Evasion and Metastatic Colonization.
- Author
-
Giordano C, La Camera G, Gelsomino L, Barone I, Bonofiglio D, Andò S, and Catalano S
- Abstract
In recent decades, the study of exosome biology has gained growing interest, representing an active area of cancer research with many potential clinical applications. Exosomes are small lipid bilayer particles released by cells with pleiotropic functions that have been reported to regulate the complex intracellular pathway involved in all steps of breast cancer development-from initiation to progression toward a metastatic dissemination. Particularly, the role of these microvesicles has been explored in metastasis, which represents the leading cause of breast cancer morbidity and mortality worldwide. Reports highlight that the plasticity of breast cancer cells, fundamental for the establishment of distant metastasis, may be in part attributed to exosome-carried signals shared between adjacent cells and long-distance cells in the body. In the present review, we will discuss the functions of exosomes in the metastatic breast cancer process and secondary site outgrowth. The possibility to decode the exosome functions in advanced diseases may offer new opportunities for early detection, molecular targeted therapies and exosome-based therapeutics in breast cancer.
- Published
- 2020
- Full Text
- View/download PDF
27. Knockdown of Leptin Receptor Affects Macrophage Phenotype in the Tumor Microenvironment Inhibiting Breast Cancer Growth and Progression.
- Author
-
Gelsomino L, Naimo GD, Malivindi R, Augimeri G, Panza S, Giordano C, Barone I, Bonofiglio D, Mauro L, Catalano S, and Andò S
- Abstract
Aberrant leptin (Ob) signaling, a hallmark of obesity, has been recognized to influence breast cancer (BC) biology within the tumor microenvironment (TME). Here, we evaluated the impact of leptin receptor (ObR) knockdown in affecting BC phenotype and in mediating the interaction between tumor cells and macrophages, the most abundant immune cells within the TME. The stable knockdown of ObR (ObR sh) in ERα-positive and ERα-negative BC cells turned the tumor phenotype into a less aggressive one, as evidenced by in vitro and in vivo models. In xenograft tumors and in co-culture experiments between circulating monocytes and BC cells, the absence of ObR reduced the recruitment of macrophages, and also affected their cytokine mRNA expression profile. This was associated with a decreased expression and secretion of monocyte chemoattractant protein-1 in ObR sh clones. The loss of Ob/ObR signaling modulated the immunosuppressive TME, as shown by a reduced expression of programmed death ligand 1/programmed cell death protein 1/arginase 1. In addition, we observed increased phagocytic activity of macrophages compared to control Sh clones in the presence of ObR sh-derived conditioned medium. Our findings, addressing an innovative role of ObR in modulating immune TME, may open new avenues to improve BC patient health care.
- Published
- 2020
- Full Text
- View/download PDF
28. Leptin and Notch Signaling Cooperate in Sustaining Glioblastoma Multiforme Progression.
- Author
-
Panza S, Russo U, Giordano F, Leggio A, Barone I, Bonofiglio D, Gelsomino L, Malivindi R, Conforti FL, Naimo GD, Giordano C, Catalano S, and Andò S
- Subjects
- Cell Movement, Cell Proliferation, Cells, Cultured, Glioblastoma pathology, Humans, Leptin genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, Notch1 genetics, Signal Transduction, Glioblastoma metabolism, Leptin metabolism, Receptor, Notch1 metabolism
- Abstract
Glioblastoma multiforme (GBM) is the most malignant form of glioma, which represents one of the commonly occurring tumors of the central nervous system. Despite the continuous development of new clinical therapies against this malignancy, it still remains a deadly disease with very poor prognosis. Here, we demonstrated the existence of a biologically active interaction between leptin and Notch signaling pathways that sustains GBM development and progression. We found that the expression of leptin and its receptors was significantly higher in human glioblastoma cells, U-87 MG and T98G, than in a normal human glial cell line, SVG p12, and that activation of leptin signaling induced growth and motility in GBM cells. Interestingly, flow cytometry and real-time RT-PCR assays revealed that GBM cells, grown as neurospheres, displayed stem cell-like properties (CD133+) along with an enhanced expression of leptin receptors. Leptin treatment significantly increased the neurosphere forming efficiency, self-renewal capacity, and mRNA expression levels of the stemness markers CD133, Nestin, SOX2, and GFAP. Mechanistically, we evidenced a leptin-mediated upregulation of Notch 1 receptor and the activation of its downstream effectors and target molecules. Leptin-induced effects on U-87 MG and T98G cells were abrogated by the selective leptin antagonist, the peptide LDFI (Leu-Asp-Phe-Ile), as well as by the specific Notch signaling inhibitor, GSI (Gamma Secretase Inhibitor) and in the presence of a dominant-negative of mastermind-like-1. Overall, these findings demonstrate, for the first time, a functional interaction between leptin and Notch signaling in GBM, highlighting leptin/Notch crosstalk as a potential novel therapeutic target for GBM treatment.
- Published
- 2020
- Full Text
- View/download PDF
29. Novel insights into adiponectin action in breast cancer: Evidence of its mechanistic effects mediated by ERα expression.
- Author
-
Andò S, Naimo GD, Gelsomino L, Catalano S, and Mauro L
- Subjects
- Adiponectin administration & dosage, Adiponectin deficiency, Breast Neoplasms etiology, Breast Neoplasms pathology, Cell Proliferation, Estrogen Receptor alpha analysis, Estrogen Receptor alpha genetics, Female, Humans, Immune System, Obesity complications, Obesity physiopathology, Risk Factors, Signal Transduction, Adiponectin physiology, Breast Neoplasms physiopathology, Estrogen Receptor alpha physiology
- Abstract
This review describes the multifaceted effects of adiponectin on breast cancer cell signalling, tumour metabolism, and microenvironment. It is largely documented that low adiponectin levels are associated with an increased risk of breast cancer. However, it needs to be still clarified what are the extents of the decrease of local/intra-tumoural adiponectin concentrations, which promote breast tumour malignancy. Most of the anti-proliferative and pro-apoptotic effects induced by adiponectin have been obtained in breast cancer cells not expressing estrogen receptor alpha (ERα). Here, we will highlight recent findings demonstrating the mechanistic effects through which adiponectin is able to fuel genomic and non-genomic estrogen signalling, inhibiting LKB1/AMPK/mTOR/S6K pathway and switching energy balance. Therefore, it emerges that the reduced adiponectin levels in patients with obesity work to sustain tumour growth and progression in ERα-positive breast cancer cells. All this may contribute to remove the misleading paradigm that adiponectin univocally inhibits breast cancer cell growth and progression independently on ERα status. The latter concept, here clearly provided by pre-clinical studies, may have translational relevance adopting adiponectin as a potential therapeutic tool. Indeed, the interfering role of ERα on adiponectin action addresses how a separate assessment of adiponectin treatment needs to be considered in novel therapeutic strategies for ERα-positive and ERα-negative breast cancer., (© 2020 World Obesity Federation.)
- Published
- 2020
- Full Text
- View/download PDF
30. Leptin Signaling Contributes to Aromatase Inhibitor Resistant Breast Cancer Cell Growth and Activation of Macrophages.
- Author
-
Gelsomino L, Giordano C, Camera G, Sisci D, Marsico S, Campana A, Tarallo R, Rinaldi A, Fuqua S, Leggio A, Grande F, Bonofiglio D, Andò S, Barone I, and Catalano S
- Subjects
- Anastrozole pharmacology, Cell Movement drug effects, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Macrophages cytology, Macrophages drug effects, Macrophages immunology, Receptors, CXCR4 metabolism, Aromatase metabolism, Aromatase Inhibitors pharmacology, Breast Neoplasms pathology, Drug Resistance, Neoplasm, Leptin metabolism, Macrophage Activation drug effects, Signal Transduction drug effects
- Abstract
Obesity represents a risk factor for breast cancer development and therapy resistance, but the molecular players underling these links are unclear. Here, we identify a role for the obesity-cytokine leptin in sustaining aromatase inhibitor (AI) resistant growth and progression in breast cancer. Using as experimental models MCF-7 breast cancer cells surviving long-term treatment with the AI anastrozole (AnaR) and Ana-sensitive counterparts, we found that AnaR cells expressed higher levels of leptin and its receptors (ObR) along with a constitutive activation of downstream effectors. Accordingly, leptin signaling inhibition reduced only AnaR cell growth and motility, highlighting the existence of an autocrine loop in mechanisms governing drug-resistant phenotypes. In agreement with ObR overexpression, increasing doses of leptin were able to stimulate to a greater extent growth and migration in AnaR than sensitive cells. Moreover, leptin contributed to enhanced crosstalk between AnaR cells and macrophages within the tumor microenvironment. Indeed, AnaR, through leptin secretion, modulated macrophage profiles and increased macrophage motility through CXCR4 signaling, as evidenced by RNA-sequencing, real-time PCR, and immunoblotting. Reciprocally, activated macrophages increased AnaR cell growth and motility in coculture systems. In conclusion, acquired AI resistance is accompanied by the development of a leptin-driven phenotype, highlighting the potential clinical benefit of targeting this cytokine network in hormone-resistant breast cancers, especially in obese women., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
31. Interfering Role of ERα on Adiponectin Action in Breast Cancer.
- Author
-
Naimo GD, Gelsomino L, Catalano S, Mauro L, and Andò S
- Subjects
- Animals, Breast Neoplasms metabolism, Female, Humans, Adiponectin metabolism, Breast Neoplasms pathology, Estrogen Receptor alpha metabolism, Obesity physiopathology
- Abstract
Obesity is characterized by an excess of adipose tissue, due to adipocyte hypertrophy and hyperplasia. Adipose tissue is an endocrine organ producing many bioactive molecules, called adipokines. During obesity, dysfunctional adipocytes alter adipokine secretion, contributing to pathophysiology of obesity-associated diseases, including metabolic syndrome, type 2-diabetes, cardiovascular diseases and many types of malignancies. Circulating adiponectin levels are inversely correlated with BMI, thus adiponectin concentrations are lower in obese than normal-weight subjects. Many clinical investigations highlight that low adiponectin levels represent a serious risk factor in breast carcinogenesis, and are associated with the development of more aggressive phenotype. A large-scale meta-analysis suggests that BMI was positively associated with breast cancer mortality in women with ERα-positive disease, regardless menopausal status. This suggests the importance of estrogen signaling contribution in breast tumorigenesis of obese patients. It has been largely demonstrated that adiponectin exerts a protective role in ERα-negative cells, promoting anti-proliferative and pro-apoptotic effects, while controversial data have been reported in ERα-positive cells. Indeed, emerging data provide evidences that adiponectin in obese patients behave as growth factor in ERα-positive breast cancer cells. This addresses how ERα signaling interference may enhance the potential inhibitory threshold of adiponectin in ERα-positive cells. Thus, we may reasonably speculate that the relatively low adiponectin concentrations could be still not adequate to elicit, in ERα-positive breast cancer cells, the same inhibitory effects observed in ERα-negative cells. In the present review we will focus on the molecular mechanisms through which adiponectin affects breast cancer cell behavior in relationship to ERα expression., (Copyright © 2020 Naimo, Gelsomino, Catalano, Mauro and Andò.)
- Published
- 2020
- Full Text
- View/download PDF
32. Modulating Tumor-Associated Macrophage Polarization by Synthetic and Natural PPARγ Ligands as a Potential Target in Breast Cancer.
- Author
-
Gionfriddo G, Plastina P, Augimeri G, Catalano S, Giordano C, Barone I, Morelli C, Giordano F, Gelsomino L, Sisci D, Witkamp R, Andò S, van Norren K, and Bonofiglio D
- Subjects
- Breast Neoplasms pathology, Cell Differentiation drug effects, Cell Line, Culture Media, Conditioned pharmacology, Cytokines metabolism, Docosahexaenoic Acids pharmacology, Ethanolamines pharmacology, Female, Humans, Ligands, Monocytes drug effects, Monocytes pathology, Rosiglitazone pharmacology, Tumor-Associated Macrophages drug effects, Tumor-Associated Macrophages metabolism, Breast Neoplasms drug therapy, Cell Polarity drug effects, Molecular Targeted Therapy, PPAR gamma metabolism, Tumor-Associated Macrophages pathology
- Abstract
Activation of peroxisome proliferator-activated receptor gamma (PPARγ) elicits anti-proliferative effects on different tumor cells, including those derived from breast cancer. PPARγ is also expressed in several cells of the breast tumor microenvironment, among which tumor associated macrophages (TAMs) play a pivotal role in tumor progression and metastasis. We explored the ability of synthetic and natural PPARγ ligands to modulate TAM polarization. The ligands included rosiglitazone (BRL-49653), and two docosahexaenoic acid (DHA) conjugates, N -docosahexaenoyl ethanolamine (DHEA) and N -docosahexaenoyl serotonin (DHA-5-HT). Human THP-1 monocytic cells were differentiated into M0, M1 and M2 macrophages that were characterized by qRT-PCR, ELISA and western blotting. A TAM-like phenotypic state was generated by adding two different breast cancer cell conditioned media (BCC-CM) to the cultures. Macrophages exposed to BCC-CM concomitantly exhibited M1 and M2 phenotypes. Interestingly, rosiglitazone, DHEA and DHA-5-HT attenuated cytokine secretion by TAMs, and this effect was reversed by the PPARγ antagonist GW9662. Given the key role played by PPARγ in the crosstalk between cancer cells and TAMs in tumor progression, its activation via endogenous or synthetic ligands may lead to novel strategies that target both epithelial neoplastic cells and the tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
33. Phosphodiesterase 5 (PDE5) Is Highly Expressed in Cancer-Associated Fibroblasts and Enhances Breast Tumor Progression.
- Author
-
Catalano S, Panza S, Augimeri G, Giordano C, Malivindi R, Gelsomino L, Marsico S, Giordano F, Győrffy B, Bonofiglio D, Andò S, and Barone I
- Abstract
The overexpression of phosphodiesterase (PDE) 5 is frequently found in various human cancers, such as those of the breast. However, PDE5's role in the tumor microenvironment is still unknown. As PDE5 represents a high-value therapeutic target, we investigated whether the expression and function of PDE5 in breast cancer-associated fibroblasts (CAFs) may be clinically relevant to malignant progression. PDE5 expression was increased in human breast cancer stroma compared with normal stroma and was correlated to a shorter overall survival. Treatment of CAFs, isolated from breast tumor biopsies, with selective PDE5 inhibitors inhibited their proliferation, motility, and invasiveness, and negatively controlled tumor-stroma interactions in both 'in vitro' and 'in vivo' models. PDE5 stable overexpression transformed immortalized mouse embryonic fibroblasts (MEFs) towards an activated fibroblast phenotype, impacting their intrinsic characteristics and paracrine effects on breast cancer cell growth and migration through an enhanced production of the C-X-C motif chemokine 16 (CXCL16). On the other hand, CAF exposure to PDE5 inhibitors was associated with reduced CXCL16 expression and secretion. Importantly, CXCL16 levels in breast cancer stroma showed a strong correlation with PDE5 levels and poor patient outcomes. In conclusion, PDE5 is overexpressed in breast cancer stroma, enhances the tumor-stimulatory activities of fibroblasts, and impacts clinical outcomes; thus, we propose this enzyme as an attractive candidate for prognosis and a potential target for treatments in breast cancer patients., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2019
- Full Text
- View/download PDF
34. Leptin Modulates Exosome Biogenesis in Breast Cancer Cells: An Additional Mechanism in Cell-to-Cell Communication.
- Author
-
Giordano C, Gelsomino L, Barone I, Panza S, Augimeri G, Bonofiglio D, Rovito D, Naimo GD, Leggio A, Catalano S, and Andò S
- Abstract
Exosomes-small membrane vesicles secreted by both normal and malignant cells upon fusion of endosomal multivesicular bodies (MVBs) with the plasma membrane-play an important role in cell-to-cell communication. During the last decade, several reports have highlighted the involvement of these nanovesicles in many aspects of breast cancer development and progression, but the extracellular signals governing their generation in breast cancer cells have not been completely unraveled. Here, we investigated the role of the obesity hormone leptin, a well-known adipokine implicated in mammary tumorigenesis, on the mechanisms regulating exosome biogenesis and release in both estrogen receptor α (ERα)-positive MCF-7 and triple-negative MDA-MB-231 breast cancer cells. We found that leptin treatment enhanced the number of MVBs in the cytoplasm of breast cancer cells and increased the amount of exosomes released in cell conditioned media. At molecular level, leptin increased the protein expression of Tsg101-a key component of the endosomal sorting complex required for transport I (ESCRT-I)-by a post-transcriptional mechanism involving its direct interaction with the chaperone protein Hsp90. Targeting leptin signaling, by a selective leptin receptor antagonist the peptide LDFI (Leu-Asp-Phe-Ile), abrogated leptin effects on Tsg101 expression and on exosome secretion in breast cancer cells. In conclusion, our findings, identifying for the first time leptin/leptin receptor/Hsp90 axis as an important regulator of exosome generation in mammary carcinoma cells, suggest that targeting this signaling pathway might represent a novel therapeutic strategy to impair exosome secretion and interrupt the dangerous cell-to-cell communication in breast cancer.
- Published
- 2019
- Full Text
- View/download PDF
35. The Emerging Role of Adiponectin in Female Malignancies.
- Author
-
Gelsomino L, Naimo GD, Catalano S, Mauro L, and Andò S
- Subjects
- Breast Neoplasms epidemiology, Female, Humans, Obesity epidemiology, Ovarian Neoplasms epidemiology, Signal Transduction, Adiponectin metabolism, Breast Neoplasms metabolism, Obesity metabolism, Ovarian Neoplasms metabolism
- Abstract
Obesity, characterized by excess body weight, is now accepted as a hazardous health condition and an oncogenic factor. In different epidemiological studies obesity has been described as a risk factor in several malignancies. Some biological mechanisms that orchestrate obesity-cancer interaction have been discovered, although others are still not completely understood. The unbalanced secretion of biomolecules, called "adipokines", released by adipocytes strongly influences obesity-related cancer development. Among these adipokines, adiponectin exerts a critical role. Physiologically adiponectin governs glucose levels and lipid metabolism and is fundamental in the reproductive system. Low adiponectin circulating levels have been found in obese patients, in which its protective effects were lost. In this review, we summarize the epidemiological, in vivo and in vitro data in order to highlight how adiponectin may affect obesity-associated female cancers.
- Published
- 2019
- Full Text
- View/download PDF
36. Leptin Receptor as a Potential Target to Inhibit Human Testicular Seminoma Growth.
- Author
-
Panza S, Gelsomino L, Malivindi R, Rago V, Barone I, Giordano C, Giordano F, Leggio A, Comandè A, Liguori A, Aquila S, Bonofiglio D, Andò S, and Catalano S
- Subjects
- Adult, Animals, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Humans, Leptin chemistry, Male, Mice, Mice, Nude, Neoplasm Proteins metabolism, Peptides chemistry, Receptors, Leptin metabolism, Seminoma metabolism, Seminoma pathology, Testicular Neoplasms metabolism, Testicular Neoplasms pathology, Xenograft Model Antitumor Assays, Leptin pharmacokinetics, Neoplasm Proteins agonists, Peptides pharmacology, Receptors, Leptin agonists, Seminoma drug therapy, Testicular Neoplasms drug therapy
- Abstract
Although in past decades the adipokine leptin and its own receptor have been considered as significant cancer biomarkers, their potential involvement in human testicular seminoma growth and progression remains unexplored. Here, we showed that the expression of leptin and its receptor was significantly higher in human testicular seminoma compared with normal adult testis. Human seminoma cell line TCam-2 also expressed leptin along with the long and short isoforms of leptin receptor, and in response to leptin treatment showed enhanced activation of its downstream effectors. In line with these results, leptin stimulation significantly increased the proliferation and migration of TCam-2 cells. Treatment of TCam-2 cells with the peptide Leu-Asp-Phe-Ile (LDFI), a full leptin-receptor antagonist, completely reversed the leptin-mediated effects on cell growth and motility as well as reduced the expression of several leptin-induced target genes. More importantly, the in vivo xenograft experiments showed that LDFI treatment markedly decreased seminoma tumor growth. Interestingly, LDFI-treated tumors showed reduced levels of the proliferation marker Ki-67 as well as decreased expression of leptin-regulated genes. Taken together, these data identify, for the first time, leptin as a key factor able to affect testicular seminoma behavior, highlighting leptin receptor as a potential target for novel potential treatments in this type of cancer., (Copyright © 2019 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. Obesity, Leptin and Breast Cancer: Epidemiological Evidence and Proposed Mechanisms.
- Author
-
Andò S, Gelsomino L, Panza S, Giordano C, Bonofiglio D, Barone I, and Catalano S
- Abstract
The prevalence of obesity has been steadily increasing over the past few decades in several developed and developing countries, with resultant hazardous health implications. Substantial epidemiological evidence has shown that excessive adiposity strongly influences risk, prognosis, and progression of various malignancies, including breast cancer. Indeed, it is now well recognized that obesity is a complex physiologic state associated with multiple molecular changes capable of modulating the behavior of breast tumor cells as well of the surrounding microenvironment. Particularly, insulin resistance, hyperactivation of insulin-like growth factor pathways, and increased levels of estrogen due to aromatization by the adipose tissue, inflammatory cytokines, and adipokines contribute to breast cancerogenesis. Among adipokines, leptin, whose circulating levels increase proportionally to total adipose tissue mass, has been identified as a key member of the molecular network in obesity. This review summarizes the current knowledge on the epidemiological link existing between obesity and breast cancer and outlines the molecular mechanisms underlying this connection. The multifaceted role of the obesity adipokine leptin in this respect is also discussed.
- Published
- 2019
- Full Text
- View/download PDF
38. Activation of Farnesoid X Receptor impairs the tumor-promoting function of breast cancer-associated fibroblasts.
- Author
-
Barone I, Vircillo V, Giordano C, Gelsomino L, Győrffy B, Tarallo R, Rinaldi A, Bruno G, Caruso A, Romeo F, Bonofiglio D, Andò S, and Catalano S
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cancer-Associated Fibroblasts drug effects, Cancer-Associated Fibroblasts pathology, Cell Movement drug effects, Cell Movement genetics, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic genetics, Coculture Techniques, Culture Media, Conditioned metabolism, Culture Media, Conditioned pharmacology, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Regulatory Networks drug effects, Hep G2 Cells, Humans, Isoxazoles pharmacology, MCF-7 Cells, Middle Aged, Receptors, Cytoplasmic and Nuclear agonists, Receptors, Cytoplasmic and Nuclear metabolism, Tumor Microenvironment drug effects, Breast Neoplasms genetics, Cancer-Associated Fibroblasts metabolism, Receptors, Cytoplasmic and Nuclear genetics, Tumor Microenvironment genetics
- Abstract
Cancer-associated Fibroblasts (CAFs), the principal components of tumor microenvironment, play multiple role in breast cancer progression. We have previously shown an oncosuppressive role of the nuclear Farnesoid X Receptor (FXR) in mammary epithelial cancer cells, here we assessed whether FXR activation may affect CAF tumor-promoting features. We showed that FXR is expressed in human CAFs isolated from four patients and treatment with the selective FXR agonist GW4064 decreased CAF migration, stress-fiber formation and contractility. RNA-sequencing highlighted cell movement and pathways known to govern cell cytoskeleton organization and migration among the most down-regulated functions and ingenuity canonical pathways upon GW4064 treatment. FXR activation reduced expression of different secreted factors. Coculture experiments revealed a reduced growth and motility of breast cancer cells treated with conditioned-media derived from GW4064-treated CAFs. Increased FXR levels in bulk tumors correlated with a longer patient survival. Our results evidence that FXR activation inhibits tumor-stimulatory activities of CAFs by impacting their mechanical properties and their paracrine signaling repertoire, suggesting that nuclear FXR ligands, by targeting both neoplastic cells and supportive stroma, may represent a promising avenue for the future management of breast cancer., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
39. Uncoupling effects of estrogen receptor α on LKB1/AMPK interaction upon adiponectin exposure in breast cancer.
- Author
-
Mauro L, Naimo GD, Gelsomino L, Malivindi R, Bruno L, Pellegrino M, Tarallo R, Memoli D, Weisz A, Panno ML, and Andò S
- Subjects
- AMP-Activated Protein Kinase Kinases, Adipokines metabolism, Adiponectin metabolism, Adipose Tissue metabolism, Animals, Cell Line, Tumor, Cell Proliferation physiology, Disease Progression, Female, Humans, MCF-7 Cells, Mice, Mice, Nude, AMP-Activated Protein Kinases metabolism, Breast Neoplasms metabolism, Estrogen Receptor alpha metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Adipose tissue is a metabolic and endocrine organ that secretes bioactive molecules called adipocytokines. Among these, adiponectin has a crucial role in obesity-associated breast cancer. The key molecule of adiponectin signaling is AMPK, which is mainly activated by liver kinase B1 (LKB1). Here, we demonstrated that estrogen receptor-α (ERα)/LKB1 interaction may negatively interfere with the LKB1 capability to phosphorylate AMPK and inhibit its downstream signaling TSC2/mTOR/p70S6k. In adiponectin-treated MCF-7 cells, AMPK signaling was not working, resulting in its downstream target acetyl-CoA carboxylase (ACC) being still active. In contrast, in MDA-MB-231 cells, AMPK and ACC phosphorylation was enhanced by adiponectin, inhibiting lipogenesis and cell growth. Upon adiponectin, ERα signaling switched the energy balance of breast cancer cells toward a lipogenic phenotype. Therefore, adiponectin played an inhibitory role on ERα-negative cell growth and progression in vitro and in vivo. In contrast, low adiponectin levels, similar to those circulating in obese patients, acted on ERα-positive cells as a growth factor, stimulating proliferation. The latter effect was blunted in vivo by high adiponectin concentration. All this may have translational relevance, addressing how the handling of adiponectin, as a therapeutic tool in breast cancer treatment, needs to be carefully considered in ERα-positive obese patients, where circulating levels of this adipocytokine are relatively low. In other words, in ERα-positive breast cancer obese patients, higher adiponectin doses should be administered with respect to ERα-negative breast cancer, also opportunely combined with antiestrogen therapy. -Mauro, L., Naimo, G. D., Gelsomino, L., Malivindi, R., Bruno, L., Pellegrino, M., Tarallo, R., Memoli, D., Weisz, A., Panno, M. L., Andò, S. Uncoupling effects of estrogen receptor α on LKB1/AMPK interaction upon adiponectin exposure in breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
40. Erratum to: ESR1 mutations affect anti-proliferative responses to tamoxifen through enhanced cross-talk with IGF signaling.
- Author
-
Gelsomino L, Gu G, Rechoum Y, Beyer AR, Pejerrey SM, Tsimelzon A, Wang T, Huffman K, Ludlow A, Andò S, and Fuqua SAW
- Published
- 2017
- Full Text
- View/download PDF
41. ESR1 mutations affect anti-proliferative responses to tamoxifen through enhanced cross-talk with IGF signaling.
- Author
-
Gelsomino L, Gu G, Rechoum Y, Beyer AR, Pejerrey SM, Tsimelzon A, Wang T, Huffman K, Ludlow A, Andò S, and Fuqua SAW
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Models, Genetic, Receptor, IGF Type 1, Receptors, Estrogen metabolism, Receptors, Somatomedin metabolism, Signal Transduction, Tamoxifen therapeutic use, Breast Neoplasms genetics, Estrogen Receptor alpha genetics, Mutation, Receptors, Somatomedin genetics, Tamoxifen pharmacology
- Abstract
The purpose of this study was to address the role of ESR1 hormone-binding mutations in breast cancer. Soft agar anchorage-independent growth assay, Western blot, ERE reporter transactivation assay, proximity ligation assay (PLA), coimmunoprecipitation assay, silencing assay, digital droplet PCR (ddPCR), Kaplan-Meier analysis, and statistical analysis. It is now generally accepted that estrogen receptor (ESR1) mutations occur frequently in metastatic breast cancers; however, we do not yet know how to best treat these patients. We have modeled the three most frequent hormone-binding ESR1 (HBD-ESR1) mutations (Y537N, Y537S, and D538G) using stable lentiviral transduction in human breast cancer cell lines. Effects on growth were examined in response to hormonal and targeted agents, and mutation-specific changes were studied using microarray and Western blot analysis. We determined that the HBD-ESR1 mutations alter anti-proliferative effects to tamoxifen (Tam), due to cell-intrinsic changes in activation of the insulin-like growth factor receptor (IGF1R) signaling pathway and levels of PIK3R1/PIK3R3. The selective estrogen receptor degrader, fulvestrant, significantly reduced the anchorage-independent growth of ESR1 mutant-expressing cells, while combination treatments with the mTOR inhibitor everolimus, or an inhibitor blocking IGF1R, and the insulin receptor significantly enhanced anti-proliferative responses. Using digital drop (dd) PCR, we identified mutations at high frequencies ranging from 12 % for Y537N, 5 % for Y537S, and 2 % for D538G in archived primary breast tumors from women treated with adjuvant mono-tamoxifen therapy. The HBD-ESR1 mutations were not associated with recurrence-free or overall survival in response in this patient cohort and suggest that knowledge of other cell-intrinsic factors in combination with ESR1 mutation status will be needed determine anti-proliferative responses to Tam.
- Published
- 2016
- Full Text
- View/download PDF
42. Glucocorticoid Receptor as a Potential Target to Decrease Aromatase Expression and Inhibit Leydig Tumor Growth.
- Author
-
Panza S, Malivindi R, Chemi F, Rago V, Giordano C, Barone I, Bonofiglio D, Gelsomino L, Giordano F, Andò S, and Catalano S
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Heterografts, Leydig Cell Tumor drug therapy, Male, Mice, Nude, Neoplasm Transplantation, Testicular Neoplasms drug therapy, Aromatase metabolism, Aromatase Inhibitors pharmacology, Dexamethasone pharmacology, Leydig Cell Tumor pathology, Receptors, Glucocorticoid antagonists & inhibitors, Testicular Neoplasms pathology
- Abstract
Leydig cell tumors are the most frequent interstitial neoplasms of the testis with increased incidence in recent years. They are hormonally active and are considered one of the steroid-secreting tumors. Although usually benign, the malignant phenotype responds poorly to conventional chemotherapy or radiation, highlighting the need to identify new therapeutic targets for treatment. Here, we identified a novel glucocorticoid-mediated mechanism that controls cell growth in Leydig cell tumors. We found that a synthetic glucocorticoid receptor agonist, dexamethasone, reduces cell proliferation in rat Leydig tumor cells by decreasing the expression and the enzymatic activity of the estrogen-producing enzyme aromatase. This inhibitory effect relies on the ability of activated glucocorticoid receptor to regulate the aromatase gene transcriptional activity through the recruitment of nuclear receptor corepressor protein and silencing mediator of retinoid and thyroid hormone receptors to a newly identified putative glucocorticoid responsive element within the aromatase promoter II. Our in vivo studies reveal a reduction of tumor growth, after dexamethasone treatment, in animal xenografts. Tumors from dexamethasone-treated mice exhibit a decrease in the expression of the proliferation marker Ki-67 and the aromatase enzyme. Our data demonstrate that activated glucocorticoid receptor, decreasing aromatase expression, induces Leydig tumor regression both in vitro and in vivo, suggesting that glucocorticoid receptor might be a potential target for the therapy of Leydig cell tumors., (Copyright © 2016. Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
43. Activated FXR Inhibits Leptin Signaling and Counteracts Tumor-promoting Activities of Cancer-Associated Fibroblasts in Breast Malignancy.
- Author
-
Giordano C, Barone I, Vircillo V, Panza S, Malivindi R, Gelsomino L, Pellegrino M, Rago V, Mauro L, Lanzino M, Panno ML, Bonofiglio D, Catalano S, and Andò S
- Subjects
- Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Cancer-Associated Fibroblasts transplantation, Carcinogenesis drug effects, Carcinogenesis genetics, Carcinogenesis metabolism, Carcinogenesis pathology, Cell Communication, Cell Line, Tumor, Culture Media, Conditioned pharmacology, Female, Humans, Leptin genetics, Leptin metabolism, MCF-7 Cells, Mice, Nude, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear metabolism, Signal Transduction, Suppressor of Cytokine Signaling 3 Protein genetics, Suppressor of Cytokine Signaling 3 Protein metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Cancer-Associated Fibroblasts drug effects, Gene Expression Regulation, Neoplastic, Isoxazoles pharmacology, Receptors, Cytoplasmic and Nuclear agonists
- Abstract
Cancer-associated fibroblasts (CAFs), the principal components of the tumor stroma, play a central role in cancer development and progression. As an important regulator of the crosstalk between breast cancer cells and CAFs, the cytokine leptin has been associated to breast carcinogenesis. The nuclear Farnesoid X Receptor-(FXR) seems to exert an oncosuppressive role in different tumors, including breast cancer. Herein, we demonstrated, for the first time, that the synthetic FXR agonist GW4064, inhibiting leptin signaling, affects the tumor-promoting activities of CAFs in breast malignancy. GW4064 inhibited growth, motility and invasiveness induced by leptin as well as by CAF-conditioned media in different breast cancer cell lines. These effects rely on the ability of activated FXR to increase the expression of the suppressor of the cytokine signaling 3 (SOCS3) leading to inhibition of leptin-activated signaling and downregulation of leptin-target genes. In vivo xenograft studies, using MCF-7 cells alone or co-injected with CAFs, showed that GW4064 administration markedly reduced tumor growth. Interestingly, GW4064-treated tumors exhibited decreased levels of leptin-regulated proteins along with a strong staining intensity for SOCS3. Thus, FXR ligands might represent an emerging potential anti-cancer therapy able to block the tumor supportive role of activated fibroblasts within the breast microenvironment.
- Published
- 2016
- Full Text
- View/download PDF
44. Androgen receptor promotes tamoxifen agonist activity by activation of EGFR in ERα-positive breast cancer.
- Author
-
Ciupek A, Rechoum Y, Gu G, Gelsomino L, Beyer AR, Brusco L, Covington KR, Tsimelzon A, and Fuqua SA
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Drug Resistance, Neoplasm, ErbB Receptors metabolism, Female, Gene Expression, Gene Expression Profiling, Humans, MAP Kinase Signaling System drug effects, Protein Binding, Receptors, Androgen genetics, Tamoxifen therapeutic use, Transcriptional Activation, rho Guanine Nucleotide Dissociation Inhibitor alpha metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, ErbB Receptors genetics, Estrogen Receptor alpha agonists, Estrogen Receptor alpha metabolism, Gene Expression Regulation, Neoplastic drug effects, Receptors, Androgen metabolism, Tamoxifen pharmacology
- Abstract
Tamoxifen (Tam) resistance represents a significant clinical problem in estrogen receptor (ER) α-positive breast cancer. We previously showed that decreased expression of Rho guanine nucleotide dissociation inhibitor (Rho GDI) α, a negative regulator of the Rho GTPase pathway, is associated with Tam resistance. We now discover that androgen receptor (AR) is overexpressed in cells with decreased Rho GDIα and seek to determine AR's contribution to resistance. We engineered ERα-positive cell lines with stable knockdown (KD) of Rho GDIα (KD cells). Resistance mechanisms were examined using microarray profiling, protein-interaction studies, growth and reporter gene assays, and Western blot analysis combined with a specific AR antagonist and other signaling inhibitors. Tam-resistant tumors and cell lines with low Rho GDIα levels exhibited upregulated AR expression. Microarray of Rho GDIα KD cells indicated that activation of EGFR and ERα was associated with Tam treatment. When AR levels were elevated, interaction between AR and EGFR was detected. Constitutive and Tam-induced phosphorylation of EGFR and ERK1/2 was blocked by the AR antagonist Enzalutamide, suggesting that AR-mediated EGFR activation was a mechanism of resistance in these cells. Constitutive ERα phosphorylation and transcriptional activity was inhibited by Enzalutamide and the EGFR inhibitor gefitinib, demonstrating that AR-mediated EGFR signaling activated ERα. Tam exhibited agonist activity in AR overexpressing cells, stimulating ERα transcriptional activity and proliferation, which was blocked by Enzalutamide and gefitinib. We describe a novel model of AR-mediated Tam resistance through activation of EGFR signaling leading to ER activation in ERα-positive cells with low expression of Rho GDIα.
- Published
- 2015
- Full Text
- View/download PDF
45. A novel leptin antagonist peptide inhibits breast cancer growth in vitro and in vivo.
- Author
-
Catalano S, Leggio A, Barone I, De Marco R, Gelsomino L, Campana A, Malivindi R, Panza S, Giordano C, Liguori A, Bonofiglio D, Liguori A, and Andò S
- Subjects
- Amino Acid Sequence, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Cell Movement drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Humans, Immunoblotting, Leptin genetics, Leptin metabolism, MCF-7 Cells, Mitogen-Activated Protein Kinases metabolism, Oligopeptides chemistry, Phosphorylation drug effects, Polyethylene Glycols chemistry, Proto-Oncogene Proteins c-akt metabolism, Receptors, Leptin antagonists & inhibitors, Receptors, Leptin metabolism, Reverse Transcriptase Polymerase Chain Reaction, STAT3 Transcription Factor metabolism, Tumor Burden drug effects, Breast Neoplasms prevention & control, Cell Proliferation drug effects, Leptin antagonists & inhibitors, Oligopeptides pharmacology, Xenograft Model Antitumor Assays
- Abstract
The role of the obesity cytokine leptin in breast cancer progression has raised interest in interfering with leptin's actions as a valuable therapeutic strategy. Leptin interacts with its receptor through three different binding sites: I-III. Site I is crucial for the formation of an active leptin-leptin receptor complex and in its subsequent activation. Amino acids 39-42 (Leu-Asp-Phe-Ile- LDFI) were shown to contribute to leptin binding site I and their mutations in alanine resulted in muteins acting as typical antagonists. We synthesized a small peptide based on the wild-type sequence of leptin binding site I (LDFI) and evaluated its efficacy in antagonizing leptin actions in breast cancer using in vitro and in vivo experimental models. The peptide LDFI abolished the leptin-induced anchorage-dependent and -independent growth as well as the migration of ERα-positive (MCF-7) and -negative (SKBR3) breast cancer cells. These results were well correlated with a reduction in the phosphorylation levels of leptin downstream effectors, as JAK2/STAT3/AKT/MAPK. Importantly, the peptide LDFI reversed the leptin-mediated up-regulation of its gene expression, as an additional mechanism able to enhance the peptide antagonistic activity. The described effects were specific for leptin signalling, since the developed peptide was not able to antagonize the other growth factors' actions on signalling activation, proliferation and migration. Finally, we showed that the LDFI pegylated peptide markedly reduced breast tumour growth in xenograft models. The unmodified peptide LDFI acting as a full leptin antagonist could become an attractive option for breast cancer treatment, especially in obese women., (© 2015 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)
- Published
- 2015
- Full Text
- View/download PDF
46. Targeting thyroid hormone receptor beta in triple-negative breast cancer.
- Author
-
Gu G, Gelsomino L, Covington KR, Beyer AR, Wang J, Rechoum Y, Huffman K, Carstens R, Andò S, and Fuqua SA
- Subjects
- Antineoplastic Agents pharmacology, Cell Line, Tumor, Docetaxel, Doxorubicin pharmacology, Female, Gene Knockdown Techniques, Humans, MCF-7 Cells, Prognosis, Signal Transduction drug effects, Taxoids pharmacology, Triple Negative Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Thyroid Hormone Receptors beta genetics, Thyroid Hormone Receptors beta metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology
- Abstract
The purpose of this study was to discover novel nuclear receptor targets in triple-negative breast cancer. Expression microarray, Western blot, qRT-PCR analyses, MTT growth assay, soft agar anchorage-independent growth assay, TRE reporter transactivation assay, and statistical analysis were performed in this study. We performed microarray analysis using 227 triple-negative breast tumors, and clustered the tumors into five groups according to their nuclear receptor expression. Thyroid hormone receptor beta (TRβ) was one of the most differentially expressed nuclear receptors in group 5 compared to other groups. TRβ low expressing patients were associated with poor outcome. We evaluated the role of TRβ in triple-negative breast cancer cell lines representing group 5 tumors. Knockdown of TRβ increased soft agar colony and reduced sensitivity to docetaxel and doxorubicin treatment. Docetaxel or doxorubicin long-term cultured cell lines also expressed decreased TRβ protein. Microarray analysis revealed cAMP/PKA signaling was the only KEGG pathways upregulated in TRβ knockdown cells. Inhibitors of cAMP or PKA, in combination with doxorubicin further enhanced cell apoptosis and restored sensitivity to chemotherapy. TRβ-specific agonists enhanced TRβ expression, and further sensitized cells to both docetaxel and doxorubicin. Sensitization was mediated by increased apoptosis with elevated cleaved PARP and caspase 3. TRβ represents a novel nuclear receptor target in triple-negative breast cancer; low TRβ levels were associated with enhanced resistance to both docetaxel and doxorubicin treatment. TRβ-specific agonists enhance chemosensitivity to these two agents. Mechanistically enhanced cAMP/PKA signaling was associated with TRβ's effects on response to chemotherapy.
- Published
- 2015
- Full Text
- View/download PDF
47. Oldenlandia diffusa extracts exert antiproliferative and apoptotic effects on human breast cancer cells through ERα/Sp1-mediated p53 activation.
- Author
-
Gu G, Barone I, Gelsomino L, Giordano C, Bonofiglio D, Statti G, Menichini F, Catalano S, and Andò S
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Estrogen Receptor Modulators pharmacology, Estrogen Receptor alpha metabolism, Estrogens metabolism, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Immunoglobulins metabolism, Oleanolic Acid pharmacology, Promoter Regions, Genetic drug effects, Tamoxifen pharmacology, Transcriptional Activation drug effects, Transcriptional Activation genetics, Triterpenes pharmacology, Tumor Suppressor Protein p53 metabolism, Up-Regulation drug effects, Ursolic Acid, Apoptosis drug effects, Breast Neoplasms drug therapy, Estrogen Receptor alpha genetics, Immunoglobulins genetics, Oldenlandia chemistry, Plant Extracts pharmacology, Tumor Suppressor Protein p53 genetics
- Abstract
Breast cancer is the most frequent tumor and a major cause of death among women. Estrogens play a crucial role in breast tumor growth, which is the rationale for the use of hormonal antiestrogen therapies. Unfortunately, not all therapeutic modalities are efficacious and it is imperative to develop new effective antitumoral drugs. Oldenlandia diffusa (OD) is a well-known medicinal plant used to prevent and treat many disorders, especially cancers. The aim of this study was to investigate the effects of OD extracts on breast cancer cell proliferation. We observed that OD extracts strongly inhibited anchorage-dependent and -independent cell growth and induced apoptosis in estrogen receptor alpha (ERα)-positive breast cancer cells, whereas proliferation and apoptotic responses of MCF-10A normal breast epithelial cells were unaffected. Mechanistically, OD extracts enhance the tumor suppressor p53 expression as a result of an increased binding of ERα/Sp1 complex to the p53 promoter region. Finally, we isolated ursolic and oleanolic acids as the bioactive compounds able to upregulate p53 expression and inhibit breast cancer cell growth. These acids were greatly effective in reducing tamoxifen-resistant growth of a derivative MCF-7 breast cancer cell line resistant to the antiestrogen treatment. Our results evidence how OD, and its bioactive compounds, exert antiproliferative and apoptotic effects selectively in ERα-positive breast cancer cells, highlighting the potential use of these herbal extracts as breast cancer preventive and/or therapeutic agents., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
48. Leptin mediates tumor-stromal interactions that promote the invasive growth of breast cancer cells.
- Author
-
Barone I, Catalano S, Gelsomino L, Marsico S, Giordano C, Panza S, Bonofiglio D, Bossi G, Covington KR, Fuqua SA, and Andò S
- Subjects
- Cell Line, Tumor, Cell Movement, Cell Proliferation, Epidermal Growth Factor metabolism, Estrogen Receptor alpha analysis, Female, Fibroblasts physiology, Gene Expression Profiling, Humans, Neoplasm Invasiveness, Receptors, Leptin analysis, Signal Transduction, Stromal Cells physiology, Breast Neoplasms pathology, Carcinoma pathology, Leptin physiology
- Abstract
Obesity confers risks to cancer development and progression but the mechanisms underlying these risks remain unclear. In this study, we identify a role for the obesity cytokine leptin, which has been implicated previously in breast cancer development, as a determinant for the tumor-promoting activity of cancer-associated fibroblasts (CAF) in both wild-type (WT) and K303R mutant estrogen receptor-α (ERα)-expressing breast cancer cells. Human CAFs stimulated a greater increase in the proliferation and migration of breast cancer cells expressing the K303R-ERα hyperactive receptor than WT-ERα-expressing cells. A concomitant increase was seen in leptin receptor isoform expression and activation of the leptin signaling pathway in cells expressing K303R-ERα compared with WT-ERα, correlating with leptin effects on cell growth, motility, and invasiveness in mutant cells. Epidermal growth factor and other factors secreted by K303R-ERα cells stimulated CAF proliferation, migration, and subsequent leptin secretion. Moreover, K303R-ERα expression generated a leptin hypersensitive phenotype in vivo. Together, our results reveal a bidirectional cross-talk between breast cancer cells and "educated" CAFs that drives tumor progression via leptin signaling. In elucidating a mechanism that connects obesity and cancer, these findings reinforce the concept that blocking cancer-stromal cell communication may represent an effective strategy for targeted therapy of breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.