Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amedeo Vetere is active.

Publication


Featured researches published by Amedeo Vetere.


Nature Reviews Drug Discovery | 2014

Targeting the pancreatic β-cell to treat diabetes

Amedeo Vetere; Amit Choudhary; Sean M. Burns; Bridget K. Wagner

Diabetes is a leading cause of morbidity and mortality worldwide, and predicted to affect over 500 million people by 2030. However, this growing burden of disease has not been met with a comparable expansion in therapeutic options. The appreciation of the pancreatic β-cell as a central player in the pathogenesis of both type 1 and type 2 diabetes has renewed focus on ways to improve glucose homeostasis by preserving, expanding and improving the function of this key cell type. Here, we provide an overview of the latest developments in this field, with an emphasis on the most promising strategies identified to date for treating diabetes by targeting the β-cell.


Journal of Biomolecular Screening | 2012

A Human Islet Cell Culture System for High-Throughput Screening:

Deepika Walpita; Thomas Hasaka; James Spoonamore; Amedeo Vetere; Karen K. Takane; Dina Fomina-Yadlin; Nathalie Fiaschi-Taesch; Alykhan F. Shamji; Paul A. Clemons; Andrew F. Stewart; Stuart L. Schreiber; Bridget K. Wagner

A small-molecule inducer of beta-cell proliferation in human islets represents a potential regeneration strategy for treating type 1 diabetes. However, the lack of suitable human beta cell lines makes such a discovery a challenge. Here, we adapted an islet cell culture system to high-throughput screening to identify such small molecules. We prepared microtiter plates containing extracellular matrix from a human bladder carcinoma cell line. Dissociated human islets were seeded onto these plates, cultured for up to 7 days, and assessed for proliferation by simultaneous Ki67 and C-peptide immunofluorescence. Importantly, this environment preserved beta-cell physiological function, as measured by glucose-stimulated insulin secretion. Adenoviral overexpression of cdk-6 and cyclin D1, known inducers of human beta cell proliferation, was used as a positive control in our assay. This induction was inhibited by cotreatment with rapamycin, an immunosuppressant often used in islet transplantation. We then performed a pilot screen of 1280 compounds, observing some phenotypic effects on cells. This high-throughput human islet cell culture method can be used to assess various aspects of beta-cell biology on a relatively large number of compounds.


Diabetes | 2016

Inhibition of DYRK1A Stimulates Human β-Cell Proliferation

Ercument Dirice; Deepika Walpita; Amedeo Vetere; Meier Bc; Kahraman S; Jiang Hu; Dančík; Burns Sm; Gilbert Tj; Olson De; Clemons Pa; Rohit N. Kulkarni; Bridget K. Wagner

Restoring functional β-cell mass is an important therapeutic goal for both type 1 and type 2 diabetes (1). While proliferation of existing β-cells is the primary means of β-cell replacement in rodents (2), it is unclear whether a similar principle applies to humans, as human β-cells are remarkably resistant to stimulation of division (3,4). Here, we show that 5-iodotubercidin (5-IT), an annotated adenosine kinase inhibitor previously reported to increase proliferation in rodent and porcine islets (5), strongly and selectively increases human β-cell proliferation in vitro and in vivo. Remarkably, 5-IT also increased glucose-dependent insulin secretion after prolonged treatment. Kinome profiling revealed 5-IT to be a potent and selective inhibitor of the dual-specificity tyrosine phosphorylation–regulated kinase (DYRK) and cell division cycle–like kinase families. Induction of β-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Gene expression profiling in whole islets treated with 5-IT revealed induction of proliferation- and cell cycle–related genes, suggesting that true proliferation is induced by 5-IT. Furthermore, 5-IT promotes β-cell proliferation in human islets grafted under the kidney capsule of NOD-scid IL2Rgnull mice. These results point to inhibition of DYRK1A as a therapeutic strategy to increase human β-cell proliferation.


ACS Chemical Biology | 2016

An Isochemogenic Set of Inhibitors To Define the Therapeutic Potential of Histone Deacetylases in β-Cell Protection

Florence F. Wagner; Morten Lundh; Taner Kaya; Patrick McCarren; Yan-Ling Zhang; Shrikanta Chattopadhyay; Jennifer Gale; Thomas Galbo; Stewart L. Fisher; Bennett C. Meier; Amedeo Vetere; Sarah J. Richardson; Noel G. Morgan; Dan Ploug Christensen; Tamara J. Gilbert; Jacob M. Hooker; Mélanie Leroy; Deepika Walpita; Thomas Mandrup-Poulsen; Bridget K. Wagner; Edward B. Holson

Modulation of histone deacetylase (HDAC) activity has been implicated as a potential therapeutic strategy for multiple diseases. However, it has been difficult to dissect the role of individual HDACs due to a lack of selective small-molecule inhibitors. Here, we report the synthesis of a series of highly potent and isoform-selective class I HDAC inhibitors, rationally designed by exploiting minimal structural changes to the clinically experienced HDAC inhibitor CI-994. We used this toolkit of isochemogenic or chemically matched inhibitors to probe the role of class I HDACs in β-cell pathobiology and demonstrate for the first time that selective inhibition of an individual HDAC isoform retains beneficial biological activity and mitigates mechanism-based toxicities. The highly selective HDAC3 inhibitor BRD3308 suppressed pancreatic β-cell apoptosis induced by inflammatory cytokines, as expected, or now glucolipotoxic stress, and increased functional insulin release. In addition, BRD3308 had no effect on human megakaryocyte differentiation, while inhibitors of HDAC1 and 2 were toxic. Our findings demonstrate that the selective inhibition of HDAC3 represents a potential path forward as a therapy to protect pancreatic β-cells from inflammatory cytokines and nutrient overload in diabetes.


Cell Metabolism | 2015

High-Throughput Luminescent Reporter of Insulin Secretion for Discovering Regulators of Pancreatic Beta-Cell Function

Sean M. Burns; Amedeo Vetere; Deepika Walpita; Vlado Dančík; Carol Khodier; Jose R. Perez; Paul A. Clemons; Bridget K. Wagner; David Altshuler

Defects in insulin secretion play a central role in the pathogenesis of type 2 diabetes, yet the mechanisms driving beta-cell dysfunction remain poorly understood, and therapies to preserve glucose-dependent insulin release are inadequate. We report a luminescent insulin secretion assay that enables large-scale investigations of beta-cell function, created by inserting Gaussia luciferase into the C-peptide portion of proinsulin. Beta-cell lines expressing this construct cosecrete luciferase and insulin in close correlation, under both standard conditions or when stressed by cytokines, fatty acids, or ER toxins. We adapted the reporter for high-throughput assays and performed a 1,600-compound pilot screen, which identified several classes of drugs inhibiting secretion, as well as glucose-potentiated secretagogues that were confirmed to have activity in primary human islets. Requiring 40-fold less time and expense than the traditional ELISA, this assay may accelerate the identification of pathways governing insulin secretion and compounds that safely augment beta-cell function in diabetes.


PLOS ONE | 2012

GW8510 Increases Insulin Expression in Pancreatic Alpha Cells through Activation of p53 Transcriptional Activity

Dina Fomina-Yadlin; Stefan Kubicek; Amedeo Vetere; Kai Hui Hu He; Stuart L. Schreiber; Bridget K. Wagner

Background Expression of insulin in terminally differentiated non-beta cell types in the pancreas could be important to treating type-1 diabetes. Previous findings led us to hypothesize involvement of kinase inhibition in induction of insulin expression in pancreatic alpha cells. Methodology/Principal Findings Alpha (αTC1.6) cells and human islets were treated with GW8510 and other small-molecule inhibitors for up to 5 days. Alpha cells were assessed for gene- and protein-expression levels, cell-cycle status, promoter occupancy status by chromatin immunoprecipitation (ChIP), and p53-dependent transcriptional activity. GW8510, a putative CDK2 inhibitor, up-regulated insulin expression in mouse alpha cells and enhanced insulin secretion in dissociated human islets. Gene-expression profiling and gene-set enrichment analysis of GW8510-treated alpha cells suggested up-regulation of the p53 pathway. Accordingly, the compound increased p53 transcriptional activity and expression levels of p53 transcriptional targets. A predicted p53 response element in the promoter region of the mouse Ins2 gene was verified by chromatin immunoprecipitation (ChIP). Further, inhibition of Jun N-terminal kinase (JNK) and p38 kinase activities suppressed insulin induction by GW8510. Conclusions/Significance The induction of Ins2 by GW8510 occurred through p53 in a JNK- and p38-dependent manner. These results implicate p53 activity in modulation of Ins2 expression levels in pancreatic alpha cells, and point to a potential approach toward using small molecules to generate insulin in an alternative cell type.


International Journal of Endocrinology | 2012

Chemical Methods to Induce Beta-Cell Proliferation

Amedeo Vetere; Bridget K. Wagner

Pancreatic beta-cell regeneration, for example, by inducing proliferation, remains an important goal in developing effective treatments for diabetes. However, beta cells have mainly been considered quiescent. This “static” view has recently been challenged by observations of relevant physiological conditions in which metabolic stress is compensated by an increase in beta-cell mass. Understanding the molecular mechanisms underlining these process could open the possibility of developing novel small molecules to increase beta-cell mass. Several cellular cell-cycle and signaling proteins provide attractive targets for high throughput screening, and recent advances in cell culture have enabled phenotypic screening for small molecule-induced beta-cell proliferation. We present here an overview of the current trends involving small-molecule approaches to induce beta-cell regeneration by proliferation.


Cell Reports | 2015

Niche-Based Screening in Multiple Myeloma Identifies a Kinesin-5 Inhibitor with Improved Selectivity over Hematopoietic Progenitors

Shrikanta Chattopadhyay; Alison L. Stewart; Siddhartha Mukherjee; Cherrie Huang; Kimberly A. Hartwell; Peter Miller; Radhika Subramanian; Leigh C. Carmody; Rushdia Z. Yusuf; David B. Sykes; Joshiawa Paulk; Amedeo Vetere; Sonia Vallet; Loredana Santo; Diana Cirstea; Teru Hideshima; Vlado Dančík; Max Majireck; Mahmud M. Hussain; Shambhavi Singh; Ryan Quiroz; Jonathan Iaconelli; Rakesh Karmacharya; Nicola Tolliday; Paul A. Clemons; M. a. l. c. o. l. m. a. S. Moore; Alykhan F. Shamji; Benjamin L. Ebert; Todd R. Golub; Noopur Raje

Novel therapeutic approaches are urgently required for multiple myeloma (MM). We used a phenotypic screening approach using co-cultures of MM cells with bone marrow stromal cells to identify compounds that overcome stromal resistance. One such compound, BRD9876, displayed selectivity over normal hematopoietic progenitors and was discovered to be an unusual ATP non-competitive kinesin-5 (Eg5) inhibitor. A novel mutation caused resistance, suggesting a binding site distinct from known Eg5 inhibitors, and BRD9876 inhibited only microtubule-bound Eg5. Eg5 phosphorylation, which increases microtubule binding, uniquely enhanced BRD9876 activity. MM cells have greater phosphorylated Eg5 than hematopoietic cells, consistent with increased vulnerability specifically to BRD9876s mode of action. Thus, differences in Eg5-microtubule binding between malignant and normal blood cells may be exploited to treat multiple myeloma. Additional steps are required for further therapeutic development, but our results indicate that unbiased chemical biology approaches can identify therapeutic strategies unanticipated by prior knowledge of protein targets.


Journal of the American Chemical Society | 2015

Kinase-Independent Small-Molecule Inhibition of JAK-STAT Signaling.

Danny Hung-Chieh Chou; Amedeo Vetere; Amit Choudhary; Stephen S. Scully; Monica Schenone; Alicia Tang; Rachel Gomez; Sean M. Burns; Morten Lundh; Tamara Vital; Eamon Comer; Patrick W. Faloon; Vlado Dančík; Christie Ciarlo; Joshiawa Paulk; Mingji Dai; Clark Reddy; Hanshi Sun; Matthew A. Young; Nicholas J. Donato; Jacob D. Jaffe; Paul A. Clemons; Michelle Palmer; Steven A. Carr; Stuart L. Schreiber; Bridget K. Wagner

Phenotypic cell-based screening is a powerful approach to small-molecule discovery, but a major challenge of this strategy lies in determining the intracellular target and mechanism of action (MoA) for validated hits. Here, we show that the small-molecule BRD0476, a novel suppressor of pancreatic β-cell apoptosis, inhibits interferon-gamma (IFN-γ)-induced Janus kinase 2 (JAK2) and signal transducer and activation of transcription 1 (STAT1) signaling to promote β-cell survival. However, unlike common JAK-STAT pathway inhibitors, BRD0476 inhibits JAK-STAT signaling without suppressing the kinase activity of any JAK. Rather, we identified the deubiquitinase ubiquitin-specific peptidase 9X (USP9X) as an intracellular target, using a quantitative proteomic analysis in rat β cells. RNAi-mediated and CRISPR/Cas9 knockdown mimicked the effects of BRD0476, and reverse chemical genetics using a known inhibitor of USP9X blocked JAK-STAT signaling without suppressing JAK activity. Site-directed mutagenesis of a putative ubiquitination site on JAK2 mitigated BRD0476 activity, suggesting a competition between phosphorylation and ubiquitination to explain small-molecule MoA. These results demonstrate that phenotypic screening, followed by comprehensive MoA efforts, can provide novel mechanistic insights into ostensibly well-understood cell signaling pathways. Furthermore, these results uncover USP9X as a potential target for regulating JAK2 activity in cellular inflammation.


Stem Cells Translational Medicine | 2016

V-Maf Musculoaponeurotic Fibrosarcoma Oncogene Homolog A Synthetic Modified mRNA Drives Reprogramming of Human Pancreatic Duct-Derived Cells Into Insulin-Secreting Cells

Elisa Corritore; Yong Syu Lee; Valentina Pasquale; Daniela Liberati; Mei Ju Hsu; Catherine Lombard; Patrick Van Der Smissen; Amedeo Vetere; Susan Bonner-Weir; Lorenzo Piemonti; Etienne Sokal; Philippe A. Lysy

β‐Cell replacement therapy represents the most promising approach to restore β‐cell mass and glucose homeostasis in patients with type 1 diabetes. Safety and ethical issues associated with pluripotent stem cells stimulated the search for adult progenitor cells with endocrine differentiation capacities. We have already described a model for expansion and differentiation of human pancreatic duct‐derived cells (HDDCs) into insulin‐producing cells. Here we show an innovative and robust in vitro system for large‐scale production of β‐like cells from HDDCs using a nonintegrative RNA‐based reprogramming technique. Synthetic modified RNAs for pancreatic transcription factors (pancreatic duodenal homeobox 1, neurogenin3, and V‐Maf musculoaponeurotic fibrosarcoma oncogene homolog A [MAFA]) were manufactured and daily transfected in HDDCs without strongly affecting immune response and cell viability. MAFA overexpression was efficient and sufficient to induce β‐cell differentiation of HDDCs, which acquired a broad repertoire of mature β‐cell markers while downregulating characteristic epithelial‐mesenchymal transition markers. Within 7 days, MAFA‐reprogrammed HDDC populations contained 37% insulin‐positive cells and a proportion of endocrine cells expressing somatostatin and pancreatic polypeptide. Ultrastructure analysis of differentiated HDDCs showed both immature and mature insulin granules with light‐backscattering properties. Furthermore, in vitro HDDC‐derived β cells (called β‐HDDCs) secreted human insulin and C‐peptide in response to glucose, KCl, 3‐isobutyl‐1‐methylxanthine, and tolbutamide stimulation. Transplantation of β‐HDDCs into diabetic SCID‐beige mice confirmed their functional glucose‐responsive insulin secretion and their capacity to mitigate hyperglycemia. Our data describe a new, reliable, and fast procedure in adult human pancreatic cells to generate clinically relevant amounts of new β cells with potential to reverse diabetes.

Collaboration


Dive into the Amedeo Vetere's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lorenzo Piemonti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Elisa Corritore

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge