Network


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

Hotspot


Dive into the research topics where Giorgio G. Galli is active.

Publication


Featured researches published by Giorgio G. Galli.


Cell | 2014

Hippo Pathway Activity Influences Liver Cell Fate

Dean Yimlamai; Constantina Christodoulou; Giorgio G. Galli; Kilangsungla Yanger; Brian Pepe-Mooney; Basanta Gurung; Kriti Shrestha; Patrick Cahan; Ben Z. Stanger; Fernando D. Camargo

The Hippo-signaling pathway is an important regulator of cellular proliferation and organ size. However, little is known about the role of this cascade in the control of cell fate. Employing a combination of lineage tracing, clonal analysis, and organoid culture approaches, we demonstrate that Hippo pathway activity is essential for the maintenance of the differentiated hepatocyte state. Remarkably, acute inactivation of Hippo pathway signaling in vivo is sufficient to dedifferentiate, at very high efficiencies, adult hepatocytes into cells bearing progenitor characteristics. These hepatocyte-derived progenitor cells demonstrate self-renewal and engraftment capacity at the single-cell level. We also identify the NOTCH-signaling pathway as a functional important effector downstream of the Hippo transducer YAP. Our findings uncover a potent role for Hippo/YAP signaling in controlling liver cell fate and reveal an unprecedented level of phenotypic plasticity in mature hepatocytes, which has implications for the understanding and manipulation of liver regeneration.


BioEssays | 2012

PRDM proteins: Important players in differentiation and disease†

Cathrine K. Fog; Giorgio G. Galli; Anders H. Lund

The PRDM family has recently spawned considerable interest as it has been implicated in fundamental aspects of cellular differentiation and exhibits expanding ties to human diseases. The PRDMs belong to the SET domain family of histone methyltransferases, however, enzymatic activity has been determined for only few PRDMs suggesting that they act by recruiting co-factors or, more speculatively, confer methylation of non-histone targets. Several PRDM family members are deregulated in human diseases, most prominently in hematological malignancies and solid cancers, where they can act as both tumor suppressors or drivers of oncogenic processes. The molecular mechanisms have been delineated for only few PRDMs and little is known about functional redundancy within the family. Future studies should identify target genes of PRDM proteins and the protein complexes in which PRDM proteins reside to provide a more comprehensive understanding of the biological and biochemical functions of this important protein family.


PLOS Genetics | 2012

Prdm5 Regulates Collagen Gene Transcription by Association with RNA Polymerase II in Developing Bone

Giorgio G. Galli; Kristian de Lichtenberg; Matteo Carrara; Wolfgang Hans; Manuela Wuelling; Bettina Mentz; Hinke A.B. Multhaupt; Cathrine K. Fog; Klaus T. Jensen; Juri Rappsilber; Andrea Vortkamp; Les Coulton; Helmut Fuchs; Valérie Gailus-Durner; Martin Hrabě de Angelis; Raffaele Calogero; John R. Couchman; Anders H. Lund

PRDM family members are transcriptional regulators involved in tissue specific differentiation. PRDM5 has been reported to predominantly repress transcription, but a characterization of its molecular functions in a relevant biological context is lacking. We demonstrate here that Prdm5 is highly expressed in developing bones; and, by genome-wide mapping of Prdm5 occupancy in pre-osteoblastic cells, we uncover a novel and unique role for Prdm5 in targeting all mouse collagen genes as well as several SLRP proteoglycan genes. In particular, we show that Prdm5 controls both Collagen I transcription and fibrillogenesis by binding inside the Col1a1 gene body and maintaining RNA polymerase II occupancy. In vivo, Prdm5 loss results in delayed ossification involving a pronounced impairment in the assembly of fibrillar collagens. Collectively, our results define a novel role for Prdm5 in sustaining the transcriptional program necessary to the proper assembly of osteoblastic extracellular matrix.


Molecular and Cellular Biology | 2013

Genomic and Proteomic Analyses of Prdm5 Reveal Interactions with Insulator Binding Proteins in Embryonic Stem Cells

Giorgio G. Galli; Matteo Carrara; Chiara Francavilla; Kristian de Lichtenberg; J. Olsen; Raffaele Calogero; Anders H. Lund

ABSTRACT PRDM proteins belong to the SET domain protein family, which is involved in the regulation of gene expression. Although few PRDM members possess histone methyltransferase activity, the molecular mechanisms by which the other members exert transcriptional regulation remain to be delineated. In this study, we find that Prdm5 is highly expressed in mouse embryonic stem (mES) cells and exploit this cellular system to characterize molecular functions of Prdm5. By combining proteomics and next-generation sequencing technologies, we identify Prdm5 interaction partners and genomic occupancy. We demonstrate that although Prdm5 is dispensable for mES cell maintenance, it directly targets genomic regions involved in early embryonic development and affects the expression of a subset of developmental regulators during cell differentiation. Importantly, Prdm5 interacts with Ctcf, cohesin, and TFIIIC and cooccupies genomic loci. In summary, our data indicate how Prdm5 modulates transcription by interacting with factors involved in genome organization in mouse embryonic stem cells.


Oncogene | 2014

Prdm5 suppresses Apc Min -driven intestinal adenomas and regulates monoacylglycerol lipase expression.

Giorgio G. Galli; Hinke A.B. Multhaupt; Matteo Carrara; K H de Lichtenberg; I B J Christensen; D Linnemann; Eric Santoni-Rugiu; Raffaele Calogero; Anders H. Lund

PRDM proteins are tissue-specific transcription factors often deregulated in diseases, particularly in cancer where different members have been found to act as oncogenes or tumor suppressors. PRDM5 is a poorly characterized member of the PRDM family for which several studies have reported a high frequency of promoter hypermethylation in cancer types of gastrointestinal origin. We report here the characterization of Prdm5 knockout mice in the context of intestinal carcinogenesis. We demonstrate that loss of Prdm5 increases the number of adenomas throughout the murine small intestine on an ApcMin background. By using the genome-wide ChIP-seq (chromatin immunoprecipitation (ChIP) followed by DNA sequencing) and transcriptome analyses we identify loci encoding proteins involved in metabolic processes as prominent PRDM5 targets and characterize monoacylglycerol lipase (Mgll) as a direct PRDM5 target in human colon cancer cells and in Prdm5 mutant mouse intestines. Moreover, we report the downregulation of PRDM5 protein expression in human colon neoplastic lesions. In summary, our data provide the first causal link between Prdm5 loss and intestinal carcinogenesis, and uncover an extensive and novel PRDM5 target repertoire likely facilitating the tumor-suppressive functions of PRDM5.


Human Molecular Genetics | 2015

A role for repressive complexes and H3K9 di-methylation in PRDM5-associated brittle cornea syndrome

Louise F. Porter; Giorgio G. Galli; Sally Williamson; Julian N. Selley; David Knight; Nursel Elcioglu; Ali Aydin; Mustafa Elcioglu; Hanka Venselaar; Anders H. Lund; Richard Bonshek; Graeme C.M. Black; Forbes D.C. Manson

Type 2 brittle cornea syndrome (BCS2) is an inherited connective tissue disease with a devastating ocular phenotype caused by mutations in the transcription factor PR domain containing 5 (PRDM5) hypothesized to exert epigenetic effects through histone and DNA methylation. Here we investigate clinical samples, including skin fibroblasts and retinal tissue from BCS2 patients, to elucidate the epigenetic role of PRDM5 and mechanisms of its dysregulation in disease. First we report abnormal retinal vascular morphology in the eyes of two cousins with BCS2 (PRDM5 Δ exons 9-14) using immunohistochemistry, and mine data from skin fibroblast expression microarrays from patients with PRDM5 mutations p.Arg590* and Δ exons 9-14, as well as from a PRDM5 ChIP-sequencing experiment. Gene ontology analysis of dysregulated PRDM5-target genes reveals enrichment for extracellular matrix (ECM) genes supporting vascular integrity and development. Q-PCR and ChIP-qPCR confirm upregulation of critical mediators of ECM stability in vascular structures (COL13A1, COL15A1, NTN1, CDH5) in patient fibroblasts. We identify H3K9 di-methylation (H3K9me2) at these PRDM5-target genes in fibroblasts, and demonstrate that the BCS2 mutation p.Arg83Cys diminishes interaction of PRDM5 with repressive complexes, including NuRD complex protein CHD4, and the repressive chromatin interactor HP1BP3, by co-immunoprecipitation combined with mass spectrometry. We observe reduced heterochromatin protein 1 binding protein 3 (HP1BP3) staining in the retinas of two cousins lacking exons 9-14 by immunohistochemistry, and dysregulated H3K9me2 in skin fibroblasts of three patients (p.Arg590*, p.Glu134* and Δ exons 9-14) by western blotting. These findings suggest that defective interaction of PRDM5 with repressive complexes, and dysregulation of H3K9me2, play a role in PRDM5-associated disease.


The EMBO Journal | 2018

Yap regulates glucose utilization and sustains nucleotide synthesis to enable organ growth

Andrew G. Cox; Allison Tsomides; Dean Yimlamai; Katie L. Hwang; Joel B. Miesfeld; Giorgio G. Galli; Brendan H. Fowl; Michael Fort; Kimberly Y Ma; Mark R. Sullivan; Aaron M. Hosios; Erin Snay; Min Yuan; Kristin K. Brown; Evan C. Lien; Sagar Chhangawala; Matthew L. Steinhauser; John M. Asara; Yariv Houvras; Brian A. Link; Matthew G. Vander Heiden; Fernando D. Camargo; Wolfram Goessling

The Hippo pathway and its nuclear effector Yap regulate organ size and cancer formation. While many modulators of Hippo activity have been identified, little is known about the Yap target genes that mediate these growth effects. Here, we show that yap−/− mutant zebrafish exhibit defects in hepatic progenitor potential and liver growth due to impaired glucose transport and nucleotide biosynthesis. Transcriptomic and metabolomic analyses reveal that Yap regulates expression of glucose transporter glut1, causing decreased glucose uptake and use for nucleotide biosynthesis in yap−/− mutants, and impaired glucose tolerance in adults. Nucleotide supplementation improves Yap deficiency phenotypes, indicating functional importance of glucose‐fueled nucleotide biosynthesis. Yap‐regulated glut1 expression and glucose uptake are conserved in mammals, suggesting that stimulation of anabolic glucose metabolism is an evolutionarily conserved mechanism by which the Hippo pathway controls organ growth. Together, our results reveal a central role for Hippo signaling in glucose metabolic homeostasis.


Journal of Cell Biology | 2018

p190 RhoGAP promotes contact inhibition in epithelial cells by repressing YAP activity

Scott R. Frank; Clemens P. Köllmann; Phi Luong; Giorgio G. Galli; Lihua Zou; Andre Bernards; Gad Getz; Raffaele Calogero; Morten Frödin; Steen H. Hansen

ARHGAP35 encoding p190A RhoGAP is a cancer-associated gene with a mutation spectrum suggestive of a tumor-suppressor function. In this study, we demonstrate that loss of heterozygosity for ARHGAP35 occurs in human tumors. We sought to identify tumor-suppressor capacities for p190A RhoGAP (p190A) and its paralog p190B in epithelial cells. We reveal an essential role for p190A and p190B to promote contact inhibition of cell proliferation (CIP), a function that relies on RhoGAP activity. Unbiased mRNA sequencing analyses establish that p190A and p190B modulate expression of genes associated with the Hippo pathway. Accordingly, we determine that p190A and p190B induce CIP by repressing YAP–TEAD-regulated gene transcription through activation of LATS kinases and inhibition of the Rho–ROCK pathway. Finally, we demonstrate that loss of a single p190 paralog is sufficient to elicit nuclear translocation of YAP and perturb CIP in epithelial cells cultured in Matrigel. Collectively, our data reveal a novel mechanism consistent with a tumor-suppressor function for ARHGAP35.


Cancer Cell | 2014

The Hippo Transducer YAP1 Transforms Activated Satellite Cells and Is a Potent Effector of Embryonal Rhabdomyosarcoma Formation

Annie M. Tremblay; Edoardo Missiaglia; Giorgio G. Galli; Simone Hettmer; Roby Urcia; Matteo Carrara; Rebekah M Judson; Khin Thway; Gema Nadal; Joanna Selfe; Graeme I. Murray; Raffaele Calogero; Cosimo De Bari; Peter S. Zammit; Mauro Delorenzi; Amy J. Wagers; Janet Shipley; Henning Wackerhage; Fernando D. Camargo


Molecular Cell | 2015

YAP Drives Growth by Controlling Transcriptional Pause Release from Dynamic Enhancers

Giorgio G. Galli; Matteo Carrara; Wei-Chien Yuan; Christian Valdes-Quezada; Basanta Gurung; Brian Pepe-Mooney; Tinghu Zhang; Geert Geeven; Nathanael S. Gray; Wouter de Laat; Raffaele Calogero; Fernando D. Camargo

Collaboration


Dive into the Giorgio G. Galli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anders H. Lund

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dean Yimlamai

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew G. Cox

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Evan C. Lien

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

John M. Asara

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Katie L. Hwang

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge