Try a new search

Format these results:

Searched for:

person:paganm02

Total Results:

248


The Long-Lost Ligase: CRL4AMBRA1 Regulates the Stability of D-Type Cyclins

Chaikovsky, Andrea C; Sage, Julien; Pagano, Michele; Simoneschi, Daniele
D-type cyclins (cyclin D1, D2, and D3, together cyclin D) are central drivers of the cell division cycle and well-described proto-oncoproteins. Rapid turnover of cyclin D is critical for its regulation, but the underlying mechanism has remained a matter of debate. Recently, AMBRA1 was identified as the major regulator of the stability of all three D-type cyclins. AMBRA1 serves as the substrate receptor for one of ∼40 CUL4-RING E3 ubiquitin ligase (CRL4) complexes to mediate the polyubiquitylation and subsequent degradation of cyclin D. Consequently, AMBRA1 regulates cell proliferation to impact tumor growth and the cellular response to cell cycle-targeted cancer therapies. Here we discuss the findings that implicate AMBRA1 as a core member of the cell cycle machinery.
PMID: 34495753
ISSN: 1557-7430
CID: 5031122

A Novel FBXO45-Gef-H1 Axis Controls Oncogenic Signaling in B-Cell Lymphoma [Meeting Abstract]

Sahasrabuddhe, A A; Chen, X; Ma, K; Wu, R; Kapoor, R; Chhipa, R R; Zhang, X; Onder, O; Wilmore, J; Gaudette, B T; Pillai, V; Li, M M; Ahn, J -Y; Chung, F; Szankasi, P; Basrur, V; Conlon, K P; Bailey, N G; Hogaboam, C M; Rottapel, R; Kim, J; Lopez, C; Seufert, J; Schlesner, M; Siebert, R; Cancro, M P; Allman, D; Pagano, M; Staudt, L M; Lim, M S; Elenitoba-Johnson, K S J
Introduction: Diffuse large B cell lymphoma (DLBCL) is the most common form of malignant lymphoma and may arise de novo, or through transformation from a pre-existing low-grade B cell lymphoma such as follicular lymphoma (FL). However, the post-translational mechanisms and deregulated pathways underlying the pathogenesis of disease evolution are not fully understood.
Method(s): We employed integrated functional and structural genomics and mass spectrometry (MS)-driven proteomics which implicated a possible novel tumor suppressor role for a conserved E3 ubiquitin ligase FBXO45 in DLBCL pathogenesis. We generated conditional knockout mice targeting loss of Fbxo45 in germinal center (GC) B-cells using the Cg1-Cre-loxP system and an assortment of CRISPR-mediated knockouts of FBXO45 in B cell lymphoma cells (FL518, BJAB, U2932). We engineered B cell lines (BJAB, U2932) to inducibly express FLAG-tagged FBXO45 to identify candidate substrates of FBXO45 using liquid chromatography-tandem MS. In vitro biochemical and in vivo studies using a variety of genetically-modified lines in xenograft studies in immunodeficient mice were performed to validate observations from proteogenomic studies. Whole genome sequencing (WGS) and genomic copy number studies were interrogated to investigate structural alterations targeting FBXO45 in primary human lymphoma samples.
Result(s): Conditional targeting of Fbxo45 in GCB-cells in transgenic mice resulted in abnormal germinal center formation with increased number and size of germinal centers. Strikingly, targeted deletion of Fbxo45 in GCB-cells resulted in spontaneous B cell lymphomas with (22/22);100%) penetrance and none of the wild-type (WT) littermates (0/20; 0%) developed lymphoma at 24 months. Macroscopic examination revealed large tumor masses, splenomegaly, and lymphadenopathy at different anatomic locations including ileocecal junction, mesenteric, retroperitoneal and cervical lymph nodes and thymus. Next generation sequencing of immunoglobulin heavy chain genes revealed monoclonal or oligoclonal B cell populations. Using proteomic analysis of affinity-purified FBXO45-immunocomplexes and differential whole proteome analysis from GCB-cells of Fbxo45 wt/wt vs Fbxo45 fl/fl mice, we discovered that FBXO45 targets the RHO guanine exchange factor GEF-H1 for ubiquitin-mediated proteasomal degradation. FBXO45 exclusively interacts with GEF H1 among 8 F-box proteins investigated and silencing of FBXO45 using three independent shRNA and CRISPR-Cas9-mediated knockouts in B-cell lymphoma cell lines promotes RHOA and MAPK activation, B cell growth and enhances proliferation. GEF-H1 is stabilized by FBXO45 depletion and GEF-H1 ubiquitination by FBXO45 requires phosphorylation of GEF-H1. Importantly, FBXO45 depletion and expression of a GEF-H1 mutant that is unable to bind FBXO45 results in GEF-H1 stabilization, promotes hyperactivated RHO and MAPK signaling and B-cell oncogenicity in vitro and in vivo. Notably, this phenotype is reverted by co-silencing of GEF-H1. Inducible ectopic expression of FBXO45 triggers accelerated turnover of GEF H1 and decreased RHOA signaling. Genomic analyses revealed recurrent loss targeting FBXO45 in transformed DLBCL (25%), de novo DLBCL (6.6%) and FL (2.3%). In keeping with our observation of prolonged hyperactivation of pERK1/2 consequent to FBXO45 ablation, in vitro and in vivo studies using B-cell lymphoma cell lines and xenografts demonstrated increased sensitivity to pharmacologic blockade with the MAP2K1/2 (ERK1/2) inhibitor Trametinib.
Conclusion(s): Our findings define a novel FBXO45-GEF-H1-MAPK signalling axis, which plays an important role in DLBCL pathogenesis. Our studies carry implications for potential exploitation of this pathway for targeted therapies. Disclosures: Siebert: AstraZeneca: Speakers Bureau. Lim: EUSA Pharma: Honoraria.
Copyright
EMBASE:2016079441
ISSN: 0006-4971
CID: 5098732

Loss of FBXO31-mediated degradation of DUSP6 dysregulates ERK and PI3K-AKT signaling and promotes prostate tumorigenesis

Duan, Shanshan; Moro, Loredana; Qu, Rui; Simoneschi, Daniele; Cho, Hyunwoo; Jiang, Shaowen; Zhao, Huiyong; Chang, Qing; de Stanchina, Elisa; Arbini, Arnaldo A; Pagano, Michele
FBXO31 is the substrate receptor of one of many CUL1-RING ubiquitin ligase (CRL1) complexes. Here, we show that low FBXO31 mRNA levels are associated with high pre-operative prostate-specific antigen (PSA) levels and Gleason grade in human prostate cancer. Mechanistically, the ubiquitin ligase CRL1FBXO31 promotes the ubiquitylation-mediated degradation of DUSP6, a dual specificity phosphatase that dephosphorylates and inactivates the extracellular-signal-regulated kinase-1 and -2 (ERK1/2). Depletion of FBXO31 stabilizes DUSP6, suppresses ERK signaling, and activates the PI3K-AKT signaling cascade. Moreover, deletion of FBXO31 promotes tumor development in a mouse orthotopic model of prostate cancer. Treatment with BCI, a small molecule inhibitor of DUSP6, suppresses AKT activation and prevents tumor formation, suggesting that the FBXO31 tumor suppressor activity is dependent on DUSP6. Taken together, our studies highlight the relevance of the FBXO31-DUSP6 axis in the regulation of ERK- and PI3K-AKT-mediated signaling pathways, as well as its therapeutic potential in prostate cancer.
PMID: 34686346
ISSN: 2211-1247
CID: 5031132

Ubiquitin ligases in cancer: Functions and clinical potentials

Duan, Shanshan; Pagano, Michele
Ubiquitylation, a highly regulated post-translational modification, controls many cellular pathways that are critical to cell homeostasis. Ubiquitin ligases recruit substrates and promote ubiquitin transfer onto targets, inducing proteasomal degradation or non-degradative signaling. Accumulating evidence highlights the critical role of dysregulated ubiquitin ligases in processes associated with the initiation and progression of cancer. Depending on the substrate specificity and biological context, a ubiquitin ligase can act either as a tumor promoter or as a tumor suppressor. In this review, we focus on the regulatory roles of ubiquitin ligases and how perturbations of their functions contribute to cancer pathogenesis. We also briefly discuss current strategies for targeting or exploiting ubiquitin ligases for cancer therapy.
PMCID:8286310
PMID: 33974914
ISSN: 2451-9448
CID: 4964702

ULK1 inhibition overcomes compromised antigen presentation and restores antitumor immunity in LKB1 mutant lung cancer

Deng, Jiehui; Thennavan, Aatish; Dolgalev, Igor; Chen, Ting; Li, Jie; Marzio, Antonio; Poirier, John T; Peng, David; Bulatovic, Mirna; Mukhopadhyay, Subhadip; Silver, Heather; Papadopoulos, Eleni; Pyon, Val; Thakurdin, Cassandra; Han, Han; Li, Fei; Li, Shuai; Ding, Hailin; Hu, Hai; Pan, Yuanwang; Weerasekara, Vajira; Jiang, Baishan; Wang, Eric S; Ahearn, Ian; Philips, Mark; Papagiannakopoulos, Thales; Tsirigos, Aristotelis; Rothenberg, Eli; Gainor, Justin; Freeman, Gordon J; Rudin, Charles M; Gray, Nathanael S; Hammerman, Peter S; Pagano, Michele; Heymach, John V; Perou, Charles M; Bardeesy, Nabeel; Wong, Kwok-Kin
PMCID:8205437
PMID: 34142094
ISSN: 2662-1347
CID: 4917722

ORF10-Cullin-2-ZYG11B complex is not required for SARS-CoV-2 infection

Mena, Elijah L; Donahue, Callie J; Vaites, Laura Pontano; Li, Jie; Rona, Gergely; O'Leary, Colin; Lignitto, Luca; Miwatani-Minter, Bearach; Paulo, Joao A; Dhabaria, Avantika; Ueberheide, Beatrix; Gygi, Steven P; Pagano, Michele; Harper, J Wade; Davey, Robert A; Elledge, Stephen J
In order to understand the transmission and virulence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is necessary to understand the functions of each of the gene products encoded in the viral genome. One feature of the SARS-CoV-2 genome that is not present in related, common coronaviruses is ORF10, a putative 38-amino acid protein-coding gene. Proteomic studies found that ORF10 binds to an E3 ubiquitin ligase containing Cullin-2, Rbx1, Elongin B, Elongin C, and ZYG11B (CRL2ZYG11B). Since CRL2ZYG11B mediates protein degradation, one possible role for ORF10 is to "hijack" CRL2ZYG11B in order to target cellular, antiviral proteins for ubiquitylation and subsequent proteasomal degradation. Here, we investigated whether ORF10 hijacks CRL2ZYG11B or functions in other ways, for example, as an inhibitor or substrate of CRL2ZYG11B While we confirm the ORF10-ZYG11B interaction and show that the N terminus of ORF10 is critical for it, we find no evidence that ORF10 is functioning to inhibit or hijack CRL2ZYG11B Furthermore, ZYG11B and its paralog ZER1 are dispensable for SARS-CoV-2 infection in cultured cells. We conclude that the interaction between ORF10 and CRL2ZYG11B is not relevant for SARS-CoV-2 infection in vitro.
PMID: 33827988
ISSN: 1091-6490
CID: 4839402

AMBRA1 regulates cyclin D to guard S-phase entry and genomic integrity

Maiani, Emiliano; Milletti, Giacomo; Nazio, Francesca; Holdgaard, Søs Grønbæk; Bartkova, Jirina; Rizza, Salvatore; Cianfanelli, Valentina; Lorente, Mar; Simoneschi, Daniele; Di Marco, Miriam; D'Acunzo, Pasquale; Di Leo, Luca; Rasmussen, Rikke; Montagna, Costanza; Raciti, Marilena; De Stefanis, Cristiano; Gabicagogeascoa, Estibaliz; Rona, Gergely; Salvador, Nélida; Pupo, Emanuela; Merchut-Maya, Joanna Maria; Daniel, Colin J; Carinci, Marianna; Cesarini, Valeriana; O'sullivan, Alfie; Jeong, Yeon-Tae; Bordi, Matteo; Russo, Francesco; Campello, Silvia; Gallo, Angela; Filomeni, Giuseppe; Lanzetti, Letizia; Sears, Rosalie C; Hamerlik, Petra; Bartolazzi, Armando; Hynds, Robert E; Pearce, David R; Swanton, Charles; Pagano, Michele; Velasco, Guillermo; Papaleo, Elena; De Zio, Daniela; Maya-Mendoza, Apolinar; Locatelli, Franco; Bartek, Jiri; Cecconi, Francesco
Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.
PMID: 33854232
ISSN: 1476-4687
CID: 4846182

CRL4AMBRA1 is a master regulator of D-type cyclins

Simoneschi, Daniele; Rona, Gergely; Zhou, Nan; Jeong, Yeon-Tae; Jiang, Shaowen; Milletti, Giacomo; Arbini, Arnaldo A; O'Sullivan, Alfie; Wang, Andrew A; Nithikasem, Sorasicha; Keegan, Sarah; Siu, Yik; Cianfanelli, Valentina; Maiani, Emiliano; Nazio, Francesca; Cecconi, Francesco; Boccalatte, Francesco; Fenyö, David; Jones, Drew R; Busino, Luca; Pagano, Michele
D-type cyclins are central regulators of the cell division cycle and are among the most frequently deregulated therapeutic targets in human cancer1, but the mechanisms that regulate their turnover are still being debated2,3. Here, by combining biochemical and genetics studies in somatic cells, we identify CRL4AMBRA1 (also known as CRL4DCAF3) as the ubiquitin ligase that targets all three D-type cyclins for degradation. During development, loss of Ambra1 induces the accumulation of D-type cyclins and retinoblastoma (RB) hyperphosphorylation and hyperproliferation, and results in defects of the nervous system that are reduced by treating pregnant mice with the FDA-approved CDK4 and CDK6 (CDK4/6) inhibitor abemaciclib. Moreover, AMBRA1 acts as a tumour suppressor in mouse models and low AMBRA1 mRNA levels are predictive of poor survival in cancer patients. Cancer hotspot mutations in D-type cyclins abrogate their binding to AMBRA1 and induce their stabilization. Finally, a whole-genome, CRISPR-Cas9 screen identified AMBRA1 as a regulator of the response to CDK4/6 inhibition. Loss of AMBRA1 reduces sensitivity to CDK4/6 inhibitors by promoting the formation of complexes of D-type cyclins with CDK2. Collectively, our results reveal the molecular mechanism that controls the stability of D-type cyclins during cell-cycle progression, in development and in human cancer, and implicate AMBRA1 as a critical regulator of the RB pathway.
PMID: 33854235
ISSN: 1476-4687
CID: 4846192

Linking ubiquitin to actin dynamics during cell fusion

Lignitto, Luca; Pagano, Michele
Cell-cell fusion is essential to the development of multicellular organisms and is driven by remodeling of the actin cytoskeleton. In this issue of Developmental Cell, Rodríguez-Pérez et al. reveal how CRL3-dependent mono-ubiquitylation modulates cell fusion by controlling the dynamics of cytoskeletal rearrangements.
PMID: 33689766
ISSN: 1878-1551
CID: 4835902

Discriminative SKP2 Interactions with CDK-Cyclin Complexes Support a Cyclin A-Specific Role in p27KIP1 Degradation

Salamina, Marco; Montefiore, Bailey C; Liu, Mengxi; Wood, Daniel J; Heath, Richard; Ault, James R; Wang, Lan-Zhen; Korolchuk, Svitlana; Baslé, Arnaud; Pastok, Martyna W; Reeks, Judith; Tatum, Natalie J; Sobott, Frank; Arold, Stefan T; Pagano, Michele; Noble, Martin E M; Endicott, Jane A
The SCFSKP2 ubiquitin ligase relieves G1 checkpoint control of CDK-cyclin complexes by promoting p27KIP1 degradation. We describe reconstitution of stable complexes containing SKP1-SKP2 and CDK1-cyclin B or CDK2-cyclin A/E, mediated by the CDK regulatory subunit CKS1. We further show that a direct interaction between a SKP2 N-terminal motif and cyclin A can stabilize SKP1-SKP2-CDK2-cyclin A complexes in the absence of CKS1. We identify the SKP2 binding site on cyclin A and demonstrate the site is not present in cyclin B or cyclin E. This site is distinct from but overlapping with features that mediate binding of p27KIP1 and other G1 cyclin regulators to cyclin A. We propose that the capacity of SKP2 to engage with CDK2-cyclin A by more than one structural mechanism provides a way to fine tune the degradation of p27KIP1 and distinguishes cyclin A from other G1 cyclins to ensure orderly cell cycle progression.
PMID: 33422522
ISSN: 1089-8638
CID: 4798592