1
|
Morin G, Garneau AP, Bouzakher N, Ségot L, Fraissenon A, Blondel A, Petit F, Chopinet C, Mayeux F, Fayoux P, Dompmartin A, Bodemer C, Balducci E, Kaltenbach S, Villarese P, Asnafi V, Legendre C, Broissand C, Fraitag S, Quelin C, Goudin N, Guibaud L, Canaud G. Somatic PIK3R1 mutations in the iSH2 domain are accessible to PI3Kα inhibition. EMBO Mol Med 2025:10.1038/s44321-025-00249-9. [PMID: 40389643 DOI: 10.1038/s44321-025-00249-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/21/2025] Open
Abstract
Mutations in PIK3R1 have recently been identified in patients with overgrowth syndromes and complex vascular malformations. PIK3R1 encodes p85α which acts as the regulatory subunit of the lipid kinase PI3Kα. PIK3R1 mutations result in the excessive activation of the AKT/mTOR pathway. Currently, there are no approved treatments specifically dedicated to patients with PIK3R1 mutations, and medical care primarily focuses on managing symptoms. In this study, we identified three patients, including two children, who had mosaic somatic PIK3R1 mutations affecting the iSH2 domain, along with severe associated symptoms that were unsuccessfully treated with rapamycin. We conducted in vitro experiments to investigate the impact of these mutations, including a double PIK3R1 mutation in cis observed in one patient. Our findings revealed that p85α mutants in the iSH2 domain showed sensitivity to alpelisib, a pharmacological inhibitor of PI3Kα. Based on these findings, we received authorization to administer alpelisib to all three patients. Following drug introduction, patients rapidly demonstrated clinical improvement, pain, fatigue and inflammatory flares were attenuated. Magnetic Resonance Imaging showed a mean decrease of 22.67% in the volume of vascular malformations over twelve months of treatment with alpelisib. No drug-related adverse events were reported during the course of the study. In conclusion, this study provides support for the use of PI3Kα inhibition as a promising therapeutic approach for individuals with PIK3R1-related anomalies.
Collapse
Affiliation(s)
- Gabriel Morin
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Alexandre P Garneau
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Nabiha Bouzakher
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Louise Ségot
- Polytech Nice Sophia, Université Côte d'Azur, Nice, France
| | - Antoine Fraissenon
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
- CREATIS UMR 5220, Villeurbanne, 69100, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
| | - Amélie Blondel
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Florence Petit
- Clinique de Génétique, CHU de Lille, Lille, 59000, France
| | - Caroline Chopinet
- Service de Physiologie & Explorations Fonctionnelles Cardiovasculaires, CHU de Lille, Lille, 59000, France
| | - Franck Mayeux
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | | | - Anne Dompmartin
- Service de Dermatologie, CHU Côte de Nacre, Caen, 14033, France
| | - Christine Bodemer
- Université Paris Cité, Paris, France
- Service de Dermatologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Estelle Balducci
- Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Sophie Kaltenbach
- Université Paris Cité, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Patrick Villarese
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Vahid Asnafi
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Laboratoire d'Oncohématologie, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Christophe Legendre
- Université Paris Cité, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | - Sylvie Fraitag
- Service d'Anatomie Pathologique, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Chloé Quelin
- Service de Génétique Clinique, Centre de Référence Anomalies du Développement de l'Ouest, CHU Rennes, Rennes, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, INSERM US24,-CNRS UAR 3633, Institut Necker-Enfants Malades, Paris, France
| | - Laurent Guibaud
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France
- Service d'Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, HCL, Bron, France
| | - Guillaume Canaud
- Université Paris Cité, Paris, France.
- INSERM U1151, Institut Necker-Enfants Malades, Paris, France.
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
| |
Collapse
|
2
|
Boff MO, Xavier FAC, Diz FM, Gonçalves JB, Ferreira LM, Zambeli J, Pazzin DB, Previato TTR, Erwig HS, Gonçalves JIB, Bruzzo FTK, Marinowic D, da Costa JC, Zanirati G. mTORopathies in Epilepsy and Neurodevelopmental Disorders: The Future of Therapeutics and the Role of Gene Editing. Cells 2025; 14:662. [PMID: 40358185 PMCID: PMC12071303 DOI: 10.3390/cells14090662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 05/15/2025] Open
Abstract
mTORopathies represent a group of neurodevelopmental disorders linked to dysregulated mTOR signaling, resulting in conditions such as tuberous sclerosis complex, focal cortical dysplasia, hemimegalencephaly, and Smith-Kingsmore Syndrome. These disorders often manifest with epilepsy, cognitive impairments, and, in some cases, structural brain anomalies. The mTOR pathway, a central regulator of cell growth and metabolism, plays a crucial role in brain development, where its hyperactivation leads to abnormal neuroplasticity, tumor formation, and heightened neuronal excitability. Current treatments primarily rely on mTOR inhibitors, such as rapamycin, which reduce seizure frequency and tumor size but fail to address underlying genetic causes. Advances in gene editing, particularly via CRISPR/Cas9, offer promising avenues for precision therapies targeting the genetic mutations driving mTORopathies. New delivery systems, including viral and non-viral vectors, aim to enhance the specificity and efficacy of these therapies, potentially transforming the management of these disorders. While gene editing holds curative potential, challenges remain concerning delivery, long-term safety, and ethical considerations. Continued research into mTOR mechanisms and innovative gene therapies may pave the way for transformative, personalized treatments for patients affected by these complex neurodevelopmental conditions.
Collapse
Affiliation(s)
- Marina Ottmann Boff
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Mendonça Diz
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Laura Meireles Ferreira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jean Zambeli
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, University of the Valley of the Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, RS, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Fernanda Thays Konat Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| |
Collapse
|
3
|
Rosell A, Krygowska AA, Alcón Pérez M, Cuesta C, Voisin MB, de Paz J, Sanz-Fraile H, Rajeeve V, Carreras-González A, Berral-González A, Swinyard O, Gabandé-Rodríguez E, Downward J, Alcaraz J, Anguita J, García-Macías C, De Las Rivas J, Cutillas PR, Castellano Sanchez E. RAS-p110α signalling in macrophages is required for effective inflammatory response and resolution of inflammation. eLife 2025; 13:RP94590. [PMID: 40272400 PMCID: PMC12021417 DOI: 10.7554/elife.94590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
Macrophages are crucial in the body's inflammatory response, with tightly regulated functions for optimal immune system performance. Our study reveals that the RAS-p110α signalling pathway, known for its involvement in various biological processes and tumourigenesis, regulates two vital aspects of the inflammatory response in macrophages: the initial monocyte movement and later-stage lysosomal function. Disrupting this pathway, either in a mouse model or through drug intervention, hampers the inflammatory response, leading to delayed resolution and the development of more severe acute inflammatory reactions in live models. This discovery uncovers a previously unknown role of the p110α isoform in immune regulation within macrophages, offering insight into the complex mechanisms governing their function during inflammation and opening new avenues for modulating inflammatory responses.
Collapse
Affiliation(s)
- Alejandro Rosell
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Agata Adelajda Krygowska
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Marta Alcón Pérez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Cristina Cuesta
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Mathieu-Benoit Voisin
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Juan de Paz
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
| | - Héctor Sanz-Fraile
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
| | - Vinothini Rajeeve
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Ana Carreras-González
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | | | - Ottilie Swinyard
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Enrique Gabandé-Rodríguez
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick InstituteLondonUnited Kingdom
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST)BarcelonaSpain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNEDerioSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Carmen García-Macías
- Pathology Unit, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de SalamancaSalamancaSpain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de SalamancaSalamancaSpain
| | - Pedro R Cutillas
- Centre for Cancer Genomics and Computational Biology, Cell Signalling and Proteomics Laboratory, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| | - Esther Castellano Sanchez
- Tumour-Stroma Signalling Lab., Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de UnamunoSalamancaSpain
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary University of LondonLondonUnited Kingdom
| |
Collapse
|
4
|
Bozich ER, Guo X, Wilson JL, Hoffmann A. A computational workflow for assessing drug effects on temporal signaling dynamics reveals robustness in stimulus-specific NFκB signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645599. [PMID: 40236106 PMCID: PMC11996442 DOI: 10.1101/2025.03.31.645599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Single-cell studies of signal transduction have revealed complex temporal dynamics that determine downstream biological function. For example, the stimulus-specific dynamics of the transcription factor NFκB specify stimulus-specific gene expression programs, and loss of specificity leads to disease. Thus, it is intriguing to consider drugs that may restore signaling specificity in disease contexts, or reduce activity but maintain signaling specificity to avoid unwanted side effects. However, while steady-state dose-response relationships have been the focus of pharmacological studies, there are no established methods for quantifying drug impact on stimulus-response signaling dynamics. Here we evaluated how drug treatments affect the stimulus-specificity of NFκB activation dynamics and its ability to accurately code ligand identity and dose. Specifically, we simulated the dynamic NFκB trajectories in response to 15 stimuli representing various immune threats under treatment of 10 representative drugs across 20 dosage levels. To quantify the effects on coding capacity, we introduced a Stimulus Response Specificity (SRS) score and a stimulus confusion score. We constructed stimulus confusion maps by employing epsilon network clustering in the trajectory space and in various dimensionally reduced spaces: canonical polyadic decomposition (CPD), functional principal component analysis (fPCA), and NFκB signaling codons (i.e. established, informative dynamic features). Our results indicated that the SRS score and the stimulus confusion map based on signaling codons are best-suited to quantify stimulus-specific NFκB dynamics confusion under pharmacological perturbations. Using these tools we found that temporal coding capacity of the NFκB signaling network is generally robust to a variety of pharmacological perturbations, thereby enabling the targeting of stimulus-specific dynamics without causing broad side-effects.
Collapse
|
5
|
Madsen RR, Le Marois A, Mruk ON, Voliotis M, Yin S, Sufi J, Qin X, Zhao SJ, Gorczynska J, Morelli D, Davidson L, Sahai E, Korolchuk VI, Tape CJ, Vanhaesebroeck B. Oncogenic PIK3CA corrupts growth factor signaling specificity. Mol Syst Biol 2025; 21:126-157. [PMID: 39706867 PMCID: PMC11791070 DOI: 10.1038/s44320-024-00078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/23/2024] Open
Abstract
Technical limitations have prevented understanding of how growth factor signals are encoded in distinct activity patterns of the phosphoinositide 3-kinase (PI3K)/AKT pathway, and how this is altered by oncogenic pathway mutations. We introduce a kinetic, single-cell framework for precise calculations of PI3K-specific information transfer for different growth factors. This features live-cell imaging of PI3K/AKT activity reporters and multiplexed CyTOF measurements of PI3K/AKT and RAS/ERK signaling markers over time. Using this framework, we found that the PIK3CAH1047R oncogene was not a simple, constitutive activator of the pathway as often presented. Dose-dependent expression of PIK3CAH1047R in human cervical cancer and induced pluripotent stem cells corrupted the fidelity of growth factor-induced information transfer, with preferential amplification of epidermal growth factor receptor (EGFR) signaling responses compared to insulin-like growth factor 1 (IGF1) and insulin receptor signaling. PIK3CAH1047R did not only shift these responses to a higher mean but also enhanced signaling heterogeneity. We conclude that oncogenic PIK3CAH1047R corrupts information transfer in a growth factor-dependent manner and suggest new opportunities for tuning of receptor-specific PI3K pathway outputs for therapeutic benefit.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK.
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| | - Alix Le Marois
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Oliwia N Mruk
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Margaritis Voliotis
- Department of Mathematics and Statistics and Living Systems Institute; University of Exeter, Exeter, EX4 4QD, UK
| | - Shaozhen Yin
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
| | - Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS, UK
| | - Salome J Zhao
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Julia Gorczynska
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Daniele Morelli
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, WC1E 6BT, UK
| | - Bart Vanhaesebroeck
- Cell Signaling Laboratory, Department of Oncology, University College London Cancer Institute Paul O'Gorman Building, University College London, London, WC1E 6BT, UK
| |
Collapse
|
6
|
Lee D, Kozurek EC, Abdullah M, Wong EJ, Li R, Liu ZS, Nguyen HD, Dickerson EB, Kim JH. PIK3CA mutation fortifies molecular determinants for immune signaling in vascular cancers. Cancer Gene Ther 2025; 32:254-267. [PMID: 39709507 PMCID: PMC11839470 DOI: 10.1038/s41417-024-00867-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Angiosarcomas are a group of vascular cancers that form malignant blood vessels. These malignancies are seemingly inflamed primarily due to their pathognomonic nature, which consists of irregular endothelium and tortuous blood channels. PIK3CA mutations are oncogenic and disrupt the PI3K pathway. In this study, we aimed to define the molecular and functional consequences of oncogenic PIK3CA mutations in angiosarcoma. We first generated two isogenic hemangiosarcoma cell lines harboring the H1047R hotspot mutations in PIK3CA gene using CRISPR/Cas9. We found PIK3CA-mutant cells established distinct molecular signatures in global gene expression and chromatin accessibility, which were associated with enrichment of immune cytokine signaling, including IL-6, IL-8, and MCP-1. These molecular processes were disrupted by the PI3K-α specific inhibitor, alpelisib. We also observed that the molecular distinctions in PIK3CA-mutant cells were linked to metabolic reprogramming in glycolytic activity and mitochondrial respiration. Our multi-omics analysis revealed that activating PIK3CA mutations regulate molecular machinery that contributes to phenotypic alterations and resistance to alpelisib. Furthermore, we identified potential therapeutic vulnerabilities of PIK3CA mutations in response to PI3K-α inhibition mediated by MAPK signaling. In summary, we demonstrate that PIK3CA mutations perpetuate PI3K activation and reinforce immune enrichment to promote drug resistance in vascular cancers.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Emma C Kozurek
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Md Abdullah
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Ethan J Wong
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Rong Li
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Zhiyan Silvia Liu
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Hai Dang Nguyen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Erin B Dickerson
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jong Hyuk Kim
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- Artificial Intelligence Academic Initiative (AI2) Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
7
|
Jashnsaz H, Neuert G. Phenotypic consequences of logarithmic signaling in MAPK stress response. iScience 2025; 28:111625. [PMID: 39886462 PMCID: PMC11780147 DOI: 10.1016/j.isci.2024.111625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 10/04/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025] Open
Abstract
How cells respond to dynamic environmental changes is crucial for understanding fundamental biological processes and cell physiology. In this study, we developed an experimental and quantitative analytical framework to explore how dynamic stress gradients that change over time regulate cellular volume, signaling activation, and growth phenotypes. Our findings reveal that gradual stress conditions substantially enhance cell growth compared to conventional acute stress. This growth advantage correlates with a minimal reduction in cell volume dependent on the dynamic of stress. We explain the growth phenotype with our finding of a logarithmic signal transduction mechanism in the yeast mitogen-activated protein kinase (MAPK) osmotic stress response pathway. These insights into the interplay between gradual environments, cell volume change, dynamic cell signaling, and growth, advance our understanding of fundamental cellular processes in gradual stress environments.
Collapse
Affiliation(s)
- Hossein Jashnsaz
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Gregor Neuert
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
8
|
Murter BM, Robinson SC, Banerjee H, Lau L, Uche UN, Szymczak-Workman AL, Kane LP. Downregulation of PIK3IP1/TrIP on T cells is controlled by TCR signal strength, PKC, and metalloprotease-mediated cleavage. J Biol Chem 2024; 300:107930. [PMID: 39454954 PMCID: PMC11615590 DOI: 10.1016/j.jbc.2024.107930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
The protein known as PI3K-interacting protein (PIK3IP1), or transmembrane inhibitor of PI3K (TrIP), is highly expressed by T cells and can modulate PI3K activity in these cells. Several studies have also revealed that TrIP is rapidly downregulated following T cell activation. However, it is unclear how this downregulation is controlled. Using a novel monoclonal antibody that robustly stains cell-surface TrIP, we demonstrate that TrIP is lost from the surface of activated T cells in a manner dependent on the strength of signaling through the T cell receptor and specific downstream signaling pathways, in particular classical PKC isoforms. TrIP expression returns by 24 h after stimulation, suggesting that it may play a role in resetting T cell receptor signaling at later time points. We also provide evidence that ADAM family proteases are required for both constitutive and stimulation-induced downregulation of TrIP in T cells. Finally, by expressing truncated forms of TrIP in cells, we identify the region in the extracellular stalk domain of TrIP that is targeted for proteolytic cleavage.
Collapse
Affiliation(s)
- Benjamin M Murter
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sean C Robinson
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Louis Lau
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Uzodinma N Uche
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
9
|
Hasegawa J. New insights into the regulation and roles of phosphatidylinositol 3,4-bisphosphate. J Biochem 2024; 176:339-345. [PMID: 39271134 DOI: 10.1093/jb/mvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024] Open
Abstract
Phosphoinositides (PIPs) are phospholipids and components of the cellular membrane. In mammals, seven phosphorylated derivatives of PIPs have been identified. Among them, phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2] is produced by lipid phosphatases (e.g., SHIP2) or by lipid kinases PI3KC2α and PI3KC2β. Although PI(3,4)P2 is undetectable in normal mouse or human tissues and common cell lines, it appears in a mouse prostate cancer model and in cells exposed to oxidative stress, indicating that PI(3,4)P2 is involved in the pathogenesis of some diseases. Here, I summarize recent findings on the cellular roles and pathophysiological significance of PI(3,4)P2.
Collapse
Affiliation(s)
- Junya Hasegawa
- Graduate School of Pharmaceutical Sciences, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| |
Collapse
|
10
|
Murter BM, Robinson SC, Banerjee H, Lau L, Uche UU, Szymczak-Workman AL, Kane LP. Downregulation of PIK3IP1/TrIP on T cells is controlled by TCR signal strength, PKC and metalloprotease-mediated cleavage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591680. [PMID: 38746242 PMCID: PMC11092459 DOI: 10.1101/2024.04.29.591680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The protein known as PI3K-interacting protein (PIK3IP1), or transmembrane inhibitor of PI3K (TrIP), is highly expressed by T cells and can modulate PI3K activity in these cells. Several studies have also revealed that TrIP is rapidly downregulated following T cell activation. However, it is unclear as to how this downregulation is controlled. Using a novel monoclonal antibody that robustly stains cell-surface TrIP, we demonstrate that TrIP is lost from the surface of activated T cells in a manner dependent on the strength of signaling through the T cell receptor (TCR) and specific downstream signaling pathways, in particular classical PKC isoforms. TrIP expression returns by 24 hours after stimulation, suggesting that it may play a role in resetting TCR signaling at later time points. We also provide evidence that ADAM family proteases are required for both constitutive and stimulation-induced downregulation of TrIP in T cells. Finally, by expressing truncated forms of TrIP in cells, we identify the region in the extracellular stalk domain of TrIP that is targeted for proteolytic cleavage.
Collapse
Affiliation(s)
- Benjamin M. Murter
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Sean C. Robinson
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Hridesh Banerjee
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Louis Lau
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Uzodinma U. Uche
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
11
|
Zheng YN, Lou SY, Lu J, Zheng FL, Tang YM, Zhang EJ, Cui SL, Zhao HJ. Selective PI3Kδ inhibitor TYM-3-98 suppresses AKT/mTOR/SREBP1-mediated lipogenesis and promotes ferroptosis in KRAS-mutant colorectal cancer. Cell Death Dis 2024; 15:474. [PMID: 38956060 PMCID: PMC11220027 DOI: 10.1038/s41419-024-06848-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most common tumors of the digestive system worldwide. KRAS mutations limit the use of anti-EGFR antibodies in combination with chemotherapy for the treatment of CRC. Therefore, novel targeted therapies are needed to overcome the KRAS-induced oncogenesis. Recent evidence suggests that inhibition of PI3K led to ferroptosis, a nonapoptotic cell death closely related to KRAS-mutant cells. Here, we showed that a selective PI3Kδ inhibitor TYM-3-98 can suppress the AKT/mTOR signaling and activate the ferroptosis pathway in KRAS-mutant CRC cells in a concentration-dependent manner. This was evidenced by the lipid peroxidation, iron accumulation, and depletion of GSH. Moreover, the overexpression of the sterol regulatory element-binding protein 1 (SREBP1), a downstream transcription factor regulating lipid metabolism, conferred CRC cells greater resistance to ferroptosis induced by TYM-3-98. In addition, the effect of TYM-3-98 was confirmed in a xenograft mouse model, which demonstrated significant tumor suppression without obvious hepatoxicity or renal toxicity. Taken together, our work demonstrated that the induction of ferroptosis contributed to the PI3Kδ inhibitor-induced cell death via the suppression of AKT/mTOR/SREBP1-mediated lipogenesis, thus displaying a promising therapeutic effect of TYM-3-98 in CRC treatment.
Collapse
Affiliation(s)
- Ya-Nan Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Si-Yue Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jun Lu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fan-Li Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yong-Mei Tang
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - En-Jun Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sun-Liang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Hua-Jun Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Mahboubi H, Yu H, Malca M, McCusty D, Stochaj U. Pifithrin-µ Induces Stress Granule Formation, Regulates Cell Survival, and Rewires Cellular Signaling. Cells 2024; 13:885. [PMID: 38891018 PMCID: PMC11172192 DOI: 10.3390/cells13110885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
(1) Background: Stress granules (SGs) are cytoplasmic protein-RNA condensates that assemble in response to various insults. SG production is driven by signaling pathways that are relevant to human disease. Compounds that modulate SG characteristics are therefore of clinical interest. Pifithrin-µ is a candidate anti-tumor agent that inhibits members of the hsp70 chaperone family. While hsp70s are required for granulostasis, the impact of pifithrin-µ on SG formation is unknown. (2) Methods: Using HeLa cells as model system, cell-based assays evaluated the effects of pifithrin-µ on cell viability. Quantitative Western blotting assessed cell signaling events and SG proteins. Confocal microscopy combined with quantitative image analyses examined multiple SG parameters. (3) Results: Pifithrin-µ induced bona fide SGs in the absence of exogenous stress. These SGs were dynamic; their properties were determined by the duration of pifithrin-µ treatment. The phosphorylation of eIF2α was mandatory to generate SGs upon pifithrin-µ exposure. Moreover, the formation of pifithrin-µ SGs was accompanied by profound changes in cell signaling. Pifithrin-µ reduced the activation of 5'-AMP-activated protein kinase, whereas the pro-survival protein kinase Akt was activated. Long-term pifithrin-µ treatment caused a marked loss of cell viability. (4) Conclusions: Our study identified stress-related changes in cellular homeostasis that are elicited by pifithrin-µ. These insights are important knowledge for the appropriate therapeutic use of pifithrin-µ and related compounds.
Collapse
Affiliation(s)
- Hicham Mahboubi
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Henry Yu
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Michael Malca
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - David McCusty
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
- Quantitative Life Sciences Program, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
13
|
Jashnsaz H, Neuert G. Phenotypic consequences of logarithmic signaling in MAPK stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570188. [PMID: 38106069 PMCID: PMC10723343 DOI: 10.1101/2023.12.05.570188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
How cells respond to dynamic environmental changes is crucial for understanding fundamental biological processes and cell physiology. In this study, we developed an experimental and quantitative analytical framework to explore how dynamic stress gradients that change over time regulate cellular volume, signaling activation, and growth phenotypes. Our findings reveal that gradual stress conditions substantially enhance cell growth compared to conventional acute stress. This growth advantage correlates with a minimal reduction in cell volume dependent on the dynamic of stress. We explain the growth phenotype with our finding of a logarithmic signal transduction mechanism in the yeast Mitogen-Activated Protein Kinase (MAPK) osmotic stress response pathway. These insights into the interplay between gradual environments, cell volume change, dynamic cell signaling, and growth, advance our understanding of fundamental cellular processes in gradual stress environments.
Collapse
Affiliation(s)
- Hossein Jashnsaz
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232 USA
| | - Gregor Neuert
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232 USA
- Lead Contact
| |
Collapse
|
14
|
Bayard C, Segna E, Taverne M, Fraissenon A, Hennocq Q, Periou B, Zerbib L, Ladraa S, Chapelle C, Hoguin C, Kaltenbach S, Villarese P, Asnafi V, Broissand C, Nemazanyy I, Autret G, Goudin N, Legendre C, Authier FJ, Viel T, Tavitian B, Gitiaux C, Fraitag S, Duong JP, Delcros C, Sergent B, Picard A, Dussiot M, Guibaud L, Khonsari R, Canaud G. Hemifacial myohyperplasia is due to somatic muscular PIK3CA gain-of-function mutations and responds to pharmacological inhibition. J Exp Med 2023; 220:e20230926. [PMID: 37712948 PMCID: PMC10503430 DOI: 10.1084/jem.20230926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 09/16/2023] Open
Abstract
Hemifacial myohyperplasia (HFMH) is a rare cause of facial asymmetry exclusively involving facial muscles. The underlying cause and the mechanism of disease progression are unknown. Here, we identified a somatic gain-of-function mutation of PIK3CA in five pediatric patients with HFMH. To understand the physiopathology of muscle hypertrophy in this context, we created a mouse model carrying specifically a PIK3CA mutation in skeletal muscles. PIK3CA gain-of-function mutation led to striated muscle cell hypertrophy, mitochondria dysfunction, and hypoglycemia with low circulating insulin levels. Alpelisib treatment, an approved PIK3CA inhibitor, was able to prevent and reduce muscle hypertrophy in the mouse model with correction of endocrine anomalies. Based on these findings, we treated the five HFMH patients. All patients demonstrated clinical, esthetical, and radiological improvement with proof of target engagement. In conclusion, we show that HFMH is due to somatic alteration of PIK3CA and is accessible to pharmacological intervention.
Collapse
Affiliation(s)
- Charles Bayard
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Eleonora Segna
- Service De Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Maxime Taverne
- Laboratoire Forme et Croissance du Crâne, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Antoine Fraissenon
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Service d’Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
- CREATIS Unité mixte de recherche 5220, Villeurbanne, France
- Service de Radiologie Mère-Enfant, Hôpital Nord, Saint Etienne, France
| | - Quentin Hennocq
- Laboratoire Forme et Croissance du Crâne, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Baptiste Periou
- Service d’anatomie Pathologique, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Lola Zerbib
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Sophia Ladraa
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
| | - Célia Chapelle
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
| | - Clément Hoguin
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Sophie Kaltenbach
- Laboratoire d’Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Patrick Villarese
- Laboratoire d’Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Vahid Asnafi
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Laboratoire d’Oncohématologie, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Christine Broissand
- Pharmacie, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Institut national de la santé et de la recherche médicale Paris, France
| | - Gwennhael Autret
- Plateforme Imageries du Vivant, Université Paris Cité, Paris Cardiovascular Research Center, Institut national de la santé et de la recherche médicale, Paris, France
| | - Nicolas Goudin
- Necker Bio-Image Analysis, Institut national de la santé et de la recherche médicale, Paris, France
| | - Christophe Legendre
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Service de Néphrologie, Transplantation Adultes, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - François-Jérôme Authier
- Service d’anatomie Pathologique, Hôpital Henri Mondor, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Thomas Viel
- Plateforme Imageries du Vivant, Université Paris Cité, Paris Cardiovascular Research Center, Institut national de la santé et de la recherche médicale, Paris, France
| | - Bertrand Tavitian
- Université Paris Cité, Paris, France
- Plateforme Imageries du Vivant, Université Paris Cité, Paris Cardiovascular Research Center, Institut national de la santé et de la recherche médicale, Paris, France
| | - Cyril Gitiaux
- Université Paris Cité, Paris, France
- Service de Neurophysiologie Clinique Pédiatrique, Centre de Référence des Pathologies Neuromusculaires, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Sylvie Fraitag
- Service d’Anatomie Pathologique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Paul Duong
- Université Paris Cité, Paris, France
- Service d’Anatomie Pathologique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Clarisse Delcros
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
| | - Bernard Sergent
- Service De Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Arnaud Picard
- Université Paris Cité, Paris, France
- Service De Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Michael Dussiot
- Institut national de la santé et de la recherche médicale U1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Laboratoire d’Excellence GR-Ex, Paris, France
| | - Laurent Guibaud
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Service d’Imagerie Pédiatrique, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Bron, France
| | - Roman Khonsari
- Université Paris Cité, Paris, France
- Service De Chirurgie Maxillo-Faciale et Chirurgie Plastique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
- Laboratoire Forme et Croissance du Crâne, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Guillaume Canaud
- Université Paris Cité, Paris, France
- Institut national de la santé et de la recherche médicale U1151, Institut Necker-Enfants Malades, Paris, France
- Unité de Médecine Translationnelle et Thérapies Ciblées, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
15
|
Kolb H, Kempf K, Martin S. Insulin and aging - a disappointing relationship. Front Endocrinol (Lausanne) 2023; 14:1261298. [PMID: 37854186 PMCID: PMC10579801 DOI: 10.3389/fendo.2023.1261298] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/25/2023] [Indexed: 10/20/2023] Open
Abstract
Experimental studies in animal models of aging such as nematodes, fruit flies or mice have observed that decreased levels of insulin or insulin signaling promotes longevity. In humans, hyperinsulinemia and concomitant insulin resistance are associated with an elevated risk of age-related diseases suggestive of a shortened healthspan. Age-related disorders include neurodegenerative diseases, hypertension, cardiovascular disease, and type 2 diabetes. High ambient insulin concentrations promote increased lipogenesis and fat storage, heightened protein synthesis and accumulation of non-functional polypeptides due to limited turnover capacity. Moreover, there is impaired autophagy activity, and less endothelial NO synthase activity. These changes are associated with mitochondrial dysfunction and oxidative stress. The cellular stress induced by anabolic activity of insulin initiates an adaptive response aiming at maintaining homeostasis, characterized by activation of the transcription factor Nrf2, of AMP activated kinase, and an unfolded protein response. This protective response is more potent in the long-lived human species than in short-lived models of aging research resulting in a stronger pro-aging impact of insulin in nematodes and fruit flies. In humans, resistance to insulin-induced cell stress decreases with age, because of an increase of insulin and insulin resistance levels but less Nrf2 activation. These detrimental changes might be contained by adopting a lifestyle that promotes low insulin/insulin resistance levels and enhances an adaptive response to cellular stress, as observed with dietary restriction or exercise.
Collapse
Affiliation(s)
- Hubert Kolb
- Faculty of Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Düsseldorf, Germany
| | - Kerstin Kempf
- West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Düsseldorf, Germany
| | - Stephan Martin
- Faculty of Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Düsseldorf, Germany
| |
Collapse
|
16
|
Madsen RR, Toker A. PI3K signaling through a biochemical systems lens. J Biol Chem 2023; 299:105224. [PMID: 37673340 PMCID: PMC10570132 DOI: 10.1016/j.jbc.2023.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
Following 3 decades of extensive research into PI3K signaling, it is now evidently clear that the underlying network does not equate to a simple ON/OFF switch. This is best illustrated by the multifaceted nature of the many diseases associated with aberrant PI3K signaling, including common cancers, metabolic disease, and rare developmental disorders. However, we are still far from a complete understanding of the fundamental control principles that govern the numerous phenotypic outputs that are elicited by activation of this well-characterized biochemical signaling network, downstream of an equally diverse set of extrinsic inputs. At its core, this is a question on the role of PI3K signaling in cellular information processing and decision making. Here, we review the determinants of accurate encoding and decoding of growth factor signals and discuss outstanding questions in the PI3K signal relay network. We emphasize the importance of quantitative biochemistry, in close integration with advances in single-cell time-resolved signaling measurements and mathematical modeling.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- MRC-Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom.
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
17
|
Chapman SP, Duprez E, Remy E. Logical modelling of myelofibrotic microenvironment predicts dysregulated progenitor stem cell crosstalk. Biosystems 2023; 231:104961. [PMID: 37392989 DOI: 10.1016/j.biosystems.2023.104961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 07/03/2023]
Abstract
Primary myelofibrosis is an untreatable age-related disorder of haematopoiesis in which a break in the crosstalk between progenitor Haematopoietic Stem Cells (HSCs) and neighbouring mesenchymal stem cells causes HSCs to rapidly proliferate and migrate out of the bone marrow. Around 90% of patients harbour mutations in driver genes that all converge to overactivate haematopoietic JAK-STAT signalling, which is thought to be critical for disease progression, as well as microenvironment modification induced by chronic inflammation. The trigger to the initial event is unknown but dysregulated thrombopoietin (TPO) and Toll-Like Receptor (TLR) signalling are hypothesised to initiate chronic inflammation which then disrupts stem cell crosstalk. Using a systems biology approach, we have constructed an intercellular logical model that captures JAK-STAT signalling and key crosstalk channels between haematopoietic and mesenchymal stem cells. The aim of the model is to decipher how TPO and TLR stimulation can perturb the bone marrow microenvironment and dysregulate stem cell crosstalk. The model predicted conditions in which the disease was averted and established for both wildtype and ectopically JAK mutated simulations. The presence of TPO and TLR are both required to disturb stem cell crosstalk and result in the disease for wildtype. TLR signalling alone was sufficient to perturb the crosstalk and drive disease progression for JAK mutated simulations. Furthermore, the model predicts probabilities of disease onset for wildtype simulations that match clinical data. These predictions might explain why patients who test negative for the JAK mutation can still be diagnosed with PMF, in which continual exposure to TPO and TLR receptor activation may trigger the initial inflammatory event that perturbs the bone marrow microenvironment and induce disease onset.
Collapse
Affiliation(s)
- S P Chapman
- I2M, Aix-Marseille University, CNRS, Marseille, France
| | - E Duprez
- Epigenetic Factors in Normal and Malignant Haematopoiesis Lab., CRCM, CNRS, INSERM, Institut Paoli Calmettes, Aix Marseille University, 13009 Marseille, France
| | - E Remy
- I2M, Aix-Marseille University, CNRS, Marseille, France.
| |
Collapse
|
18
|
Cirillo D, Diceglie M, Nazaré M. Isoform-selective targeting of PI3K: time to consider new opportunities? Trends Pharmacol Sci 2023; 44:601-621. [PMID: 37438206 DOI: 10.1016/j.tips.2023.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/14/2023]
Abstract
Phosphoinositide-3-kinases (PI3Ks) are central to several cellular signaling pathways in human physiology and are potential pharmacological targets for many pathologies including cancer, thrombosis, and pulmonary diseases. Tremendous efforts to develop isoform-selective inhibitors have culminated in the approval of several drugs, validating PI3K as a tractable and therapeutically relevant target. Although successful therapeutic validation has focused on isoform-selective class I orthosteric inhibitors, recent clinical findings have indicated challenges regarding poor drug tolerance owing to sustained on-target inhibition. Hence, additional approaches are warranted to increase the clinical benefits of specific clinical treatment options, which may involve the employment of so far underexploited targeting modalities or the development of inhibitors for currently underexplored PI3K class II isoforms. We review recent key discoveries in the development of isoform-selective inhibitors, focusing particularly on PI3K class II isoforms, and highlight the emerging importance of developing a broader arsenal of pharmacological tools.
Collapse
Affiliation(s)
- Davide Cirillo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Berlin, Germany
| | - Marta Diceglie
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Berlin, Germany
| | - Marc Nazaré
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Berlin, Germany.
| |
Collapse
|
19
|
Gong GQ, Bilanges B, Allsop B, Masson GR, Roberton V, Askwith T, Oxenford S, Madsen RR, Conduit SE, Bellini D, Fitzek M, Collier M, Najam O, He Z, Wahab B, McLaughlin SH, Chan AWE, Feierberg I, Madin A, Morelli D, Bhamra A, Vinciauskaite V, Anderson KE, Surinova S, Pinotsis N, Lopez-Guadamillas E, Wilcox M, Hooper A, Patel C, Whitehead MA, Bunney TD, Stephens LR, Hawkins PT, Katan M, Yellon DM, Davidson SM, Smith DM, Phillips JB, Angell R, Williams RL, Vanhaesebroeck B. A small-molecule PI3Kα activator for cardioprotection and neuroregeneration. Nature 2023; 618:159-168. [PMID: 37225977 PMCID: PMC7614683 DOI: 10.1038/s41586-023-05972-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/17/2023] [Indexed: 05/26/2023]
Abstract
Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.
Collapse
Affiliation(s)
- Grace Q Gong
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Benoit Bilanges
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Ben Allsop
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Glenn R Masson
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Victoria Roberton
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Trevor Askwith
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Sally Oxenford
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Ralitsa R Madsen
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Sarah E Conduit
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Dom Bellini
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Martina Fitzek
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Matt Collier
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Osman Najam
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ben Wahab
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | | | - A W Edith Chan
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Andrew Madin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Daniele Morelli
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Amandeep Bhamra
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Vanesa Vinciauskaite
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Silvia Surinova
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK
| | | | - Matthew Wilcox
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Alice Hooper
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Chandni Patel
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Maria A Whitehead
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Tom D Bunney
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | | | | | - Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - David M Smith
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Richard Angell
- Drug Discovery Group, Translational Research Office, University College London, London, UK
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Roger L Williams
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
20
|
Prinz R. Nothing in evolution makes sense except in the light of code biology. Biosystems 2023; 229:104907. [PMID: 37207840 DOI: 10.1016/j.biosystems.2023.104907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023]
Abstract
This article highlights the potential contribution of biological codes to the course and dynamics of evolution. The concept of organic codes, developed by Marcello Barbieri, has fundamentally changed our view of how living systems function. The notion that molecular interactions built on adaptors that arbitrarily link molecules from different "worlds" in a conventional, i.e., rule-based way, departs significantly from the law-based constraints imposed on livening things by physical and chemical mechanisms. In other words, living and non-living things behave like rules and laws, respectively, but this important distinction is rarely considered in current evolutionary theory. The many known codes allow quantification of codes that relate to a cell, or comparisons between different biological systems and may pave the way to a quantitative and empirical research agenda in code biology. A starting point for such an endeavour is the introduction of a simple dichotomous classification of structural and regulatory codes. This classification can be used as a tool to analyse and quantify key organising principles of the living world, such as modularity, hierarchy, and robustness, based on organic codes. The implications for evolutionary research are related to the unique dynamics of codes, or ´Eigendynamics´ (self-momentum) and how they determine the behaviour of biological systems from within, whereas physical constraints are imposed mainly from without. A speculation on the drivers of macroevolution in light of codes is followed by the conclusion that a meaningful and comprehensive understanding of evolution depends including codes into the equation of life.
Collapse
|
21
|
Lobo V, Rocha A, Castro TG, Carvalho MA. Synthesis of Novel 2,9-Disubstituted-6-morpholino Purine Derivatives Assisted by Virtual Screening and Modelling of Class I PI3K Isoforms. Polymers (Basel) 2023; 15:polym15071703. [PMID: 37050317 PMCID: PMC10096987 DOI: 10.3390/polym15071703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The phosphatidylinositol-3 kinase (PI3K) pathway is one of the most frequently activated pathogenic signalling cascades in a wide variety of cancers. In the last 15 years, there has been an increase in the search for selective inhibitors of the four class I isoforms of PI3K, as they demonstrate better specificity and reduced toxicity in comparison to existing inhibitors. A ligand-based and target-based rational drug design strategy was employed to build a virtual library of 105 new compounds. Through this strategy, the four isoforms were compared regarding their activity pocket availability, amino acid sequences, and prone interactions. Additionally, a known active scaffold was used as a molecular base to design new derivatives. The virtual screening of the resultant library toward the four isoforms points to the obtention of 19 selective inhibitors for the PI3Kα and PI3Kγ targets. Three selective ligands, one for α-isoform and two for γ-isoform, present a ∆ (∆Gbinding) equal or greater than 1.5 Kcal/mol and were identified as the most promising candidates. A principal component analysis was used to establish correlations between the affinity data and some of the physicochemical and structural properties of the ligands. The binding modes and interactions established by the selective ligands in the active centre of the α and γ isoforms of PI3K were also investigated. After modelling studies, a synthetic approach to generate selective ligands was developed and applied in synthesising a set of derivatives that were obtained in good to excellent yield.
Collapse
|
22
|
Targeting PI3K/AKT signaling pathway in obesity. Biomed Pharmacother 2023; 159:114244. [PMID: 36638594 DOI: 10.1016/j.biopha.2023.114244] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Obesity is a disorder with an increasing prevalence, which impairs the life quality of patients and intensifies societal health care costs. The development of safe and innovative prevention strategies and therapeutic approaches is thus of great importance. The complex pathophysiology of obesity involves multiple signaling pathways that influence energy metabolism in different tissues. The phosphatidylinositol 3-kinases (PI3K)/protein kinase B (AKT) pathway is critical for the metabolic homeostasis and its function in insulin-sensitive tissues is described in the context of health, obesity and obesity-related complications. The PI3K family participates in the regulation of diverse physiological processes including but not limited to cell growth, survival, differentiation, autophagy, chemotaxis, and metabolism depending on the cellular context. AKT is downstream of PI3K in the insulin signaling pathway, and promotes multiple cellular processes by targeting a plethora of regulatory proteins that control glucose and lipid metabolism. Natural products are essential for prevention and treatment of many human diseases, including obesity. Anti-obesity natural compounds effect multiple pathophysiological mechanisms involved in obesity development. Numerous recent preclinical studies reveal the advances in using plant secondary metabolites to target the PI3K/AKT signaling pathway for obesity management. In this paper the druggability of PI3K as a target for compounds with anti-obesity potential is evaluated. Perspectives on the strategies and limitations for clinical implementation of obesity management using natural compounds modulating the PI3K/AKT pathway are suggested.
Collapse
|
23
|
Targeting Class I-II-III PI3Ks in Cancer Therapy: Recent Advances in Tumor Biology and Preclinical Research. Cancers (Basel) 2023; 15:cancers15030784. [PMID: 36765741 PMCID: PMC9913247 DOI: 10.3390/cancers15030784] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
Phosphatidylinositol-3-kinase (PI3K) enzymes, producing signaling phosphoinositides at plasma and intracellular membranes, are key in intracellular signaling and vesicular trafficking pathways. PI3K is a family of eight enzymes divided into three classes with various functions in physiology and largely deregulated in cancer. Here, we will review the recent evidence obtained during the last 5 years on the roles of PI3K class I, II and III isoforms in tumor biology and on the anti-tumoral action of PI3K inhibitors in preclinical cancer models. The dependency of tumors to PI3K isoforms is dictated by both genetics and context (e.g., the microenvironment). The understanding of class II/III isoforms in cancer development and progression remains scarce. Nonetheless, the limited available data are consistent and reveal that there is an interdependency between the pathways controlled by all PI3K class members in their role to promote cancer cell proliferation, survival, growth, migration and metabolism. It is unknown whether this feature contributes to partial treatment failure with isoform-selective PI3K inhibitors. Hence, a better understanding of class II/III functions to efficiently inhibit their positive and negative interactions with class I PI3Ks is needed. This research will provide the proof-of-concept to develop combination treatment strategies targeting several PI3K isoforms simultaneously.
Collapse
|
24
|
Oncogenic mutations of PIK3CA lead to increased membrane recruitment driven by reorientation of the ABD, p85 and C-terminus. Nat Commun 2023; 14:181. [PMID: 36635288 PMCID: PMC9837058 DOI: 10.1038/s41467-023-35789-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/29/2022] [Indexed: 01/14/2023] Open
Abstract
PIK3CA encoding the phosphoinositide 3-kinase (PI3K) p110α catalytic subunit is frequently mutated in cancer, with mutations occurring widely throughout the primary sequence. The full set of mechanisms underlying how PI3Ks are activated by all oncogenic mutations on membranes are unclear. Using a synergy of biochemical assays and hydrogen deuterium exchange mass spectrometry (HDX-MS), we reveal unique regulatory mechanisms underlying PI3K activation. Engagement of p110α on membranes leads to disengagement of the ABD of p110α from the catalytic core, and the C2 domain from the iSH2 domain of the p85 regulatory subunit. PI3K activation also requires reorientation of the p110α C-terminus, with mutations that alter the inhibited conformation of the C-terminus increasing membrane binding. Mutations at the C-terminus (M1043I/L, H1047R, G1049R, and N1068KLKR) activate p110α through distinct mechanisms, with this having important implications for mutant selective inhibitor development. This work reveals unique mechanisms underlying how PI3K is activated by oncogenic mutations, and explains how double mutants can synergistically increase PI3K activity.
Collapse
|
25
|
Tibarewal P, Rathbone V, Constantinou G, Pearce W, Adil M, Varyova Z, Folkes L, Hampson A, Classen GAE, Alves A, Carvalho S, Scudamore CL, Vanhaesebroeck B. Long-term treatment of cancer-prone germline PTEN mutant mice with low-dose rapamycin extends lifespan and delays tumour development. J Pathol 2022; 258:382-394. [PMID: 36073856 PMCID: PMC9828006 DOI: 10.1002/path.6009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/19/2022] [Accepted: 09/05/2022] [Indexed: 01/19/2023]
Abstract
PTEN is one of the most commonly inactivated tumour suppressor genes in sporadic cancer. Germline heterozygous PTEN gene alterations also underlie PTEN hamartoma tumour syndrome (PHTS), a rare human cancer-predisposition condition. A key feature of systemic PTEN deregulation is the inability to adequately dampen PI3-kinase (PI3K)/mTORC1 signalling. PI3K/mTORC1 pathway inhibitors such as rapamycin are therefore expected to neutralise the impact of PTEN loss, rendering this a more druggable context compared with those of other tumour suppressor pathways such as loss of TP53. However, this has not been explored in cancer prevention in a model of germline cancer predisposition, such as PHTS. Clinical trials of short-term treatment with rapamycin have recently been initiated for PHTS, focusing on cognition and colon polyposis. Here, we administered a low dose of rapamycin from the age of 6 weeks onwards to mice with heterozygous germline Pten loss, a mouse model that recapitulates most characteristics of human PHTS. Rapamycin was well tolerated and led to a highly significant improvement of survival in both male and female mice. This was accompanied by a delay in, but not full blockade of, the development of a range of proliferative lesions, including gastro-intestinal and thyroid tumours and endometrial hyperplasia, with no impact on mammary and prostate tumours, and no effect on brain overgrowth. Our data indicate that rapamycin may have cancer prevention potential in human PHTS. This might also be the case for sporadic cancers in which genetic PI3K pathway activation is an early event in tumour development, such as endometrial cancer and some breast cancers. To the best of our knowledge, this is the first report of a long-term treatment of a germline cancer predisposition model with a PI3K/mTOR pathway inhibitor. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | | | - Wayne Pearce
- Cancer Institute, University College London, London, UK
| | - Mahreen Adil
- Cancer Institute, University College London, London, UK
| | - Zofia Varyova
- Cancer Institute, University College London, London, UK
| | - Lisa Folkes
- Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Alix Hampson
- Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | | | - Adriana Alves
- Cancer Institute, University College London, London, UK
| | - Sara Carvalho
- Cancer Institute, University College London, London, UK
| | | | | |
Collapse
|
26
|
Ezeani M, Prabhu S. PI3K signalling at the intersection of cardio-oncology networks: cardiac safety in the era of AI. Cell Mol Life Sci 2022; 79:594. [PMID: 36380172 PMCID: PMC11803020 DOI: 10.1007/s00018-022-04627-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/07/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
Class I phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases. They are super elevated in many human cancer types and exert their main cellular functions by activating Akt to trigger an array of distinct responses, affecting metabolism and cell polarity. The signal equally plays important roles in cardiovascular pathophysiology. PI3K is required for cardiogenesis and regulation of cardiac structure and function. Overexpression of PI3K governs the development of cardiac pressure overload adaptation and compensatory hypertrophy. Therefore, inhibition of PI3K shortens life span, enhances cardiac dysfunction and pathological hypertrophy. The inverse inhibition effect, however, desirably destroys many cancer cells by blocking several aspects of the tumorigenesis phenotype. Given the contrasting effects in cardio-oncology; the best therapeutic strategy to target PI3K in cancer, while maintaining or rather increasing cardiac safety is under intense investigational scrutiny. To improve our molecular understanding towards identifying cardiac safety signalling of PI3K and/or better therapeutic strategy for cancer treatment, this article reviews PI3K signalling in cardio-oncology. PI3K signalling at the interface of metabolism, inflammation and immunity, and autonomic innervation networks were examined. Examples were then given of cardiovascular drugs that target the networks, being repurposed for cancer treatment. This was followed by an intersection scheme of the networks that can be functionalised with machine learning for safety and risk prediction, diagnoses, and defining new novel encouraging leads and targets for clinical translation. This will hopefully overcome the challenges of the one-signalling-one-health-outcome alliance, and expand our knowledge of the totality of PI3K signalling in cardio-oncology.
Collapse
Affiliation(s)
- Martin Ezeani
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
| | - Sandeep Prabhu
- The Alfred, and University of Melbourne, Parkville, Melbourne, VIC, 3010, Australia
| |
Collapse
|
27
|
Angulo-Urarte A, Graupera M. When, where and which PIK3CA mutations are pathogenic in congenital disorders. NATURE CARDIOVASCULAR RESEARCH 2022; 1:700-714. [PMID: 39196083 DOI: 10.1038/s44161-022-00107-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/22/2022] [Indexed: 08/29/2024]
Abstract
PIK3CA encodes the class I PI3Kα isoform and is frequently mutated in cancer. Activating mutations in PIK3CA also cause a range of congenital disorders featuring asymmetric tissue overgrowth, known as the PIK3CA-related overgrowth spectrum (PROS), with frequent vascular involvement. In PROS, PIK3CA mutations arise postzygotically, during embryonic development, leading to a mosaic body pattern distribution resulting in a variety of phenotypic features. A clear skewed pattern of overgrowth favoring some mesoderm-derived and ectoderm-derived tissues is observed but not understood. Here, we summarize our current knowledge of the determinants of PIK3CA-related pathogenesis in PROS, including intrinsic factors such as cell lineage susceptibility and PIK3CA variant bias, and extrinsic factors, which refers to environmental modifiers. We also include a section on PIK3CA-related vascular malformations given that the vasculature is frequently affected in PROS. Increasing our biological understanding of PIK3CA mutations in PROS will contribute toward unraveling the onset and progression of these conditions and ultimately impact on their treatment. Given that PIK3CA mutations are similar in PROS and cancer, deeper insights into one will also inform about the other.
Collapse
Affiliation(s)
- Ana Angulo-Urarte
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
28
|
Vasan N, Cantley LC. At a crossroads: how to translate the roles of PI3K in oncogenic and metabolic signalling into improvements in cancer therapy. Nat Rev Clin Oncol 2022; 19:471-485. [PMID: 35484287 PMCID: PMC11215755 DOI: 10.1038/s41571-022-00633-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 12/14/2022]
Abstract
Numerous agents targeting various phosphatidylinositol 3-kinase (PI3K) pathway components, including PI3K, AKT and mTOR, have been tested in oncology clinical trials, resulting in regulatory approvals for the treatment of selected patients with breast cancer, certain other solid tumours or particular haematological malignancies. However, given the prominence of PI3K signalling in cancer and the crucial role of this pathway in linking cancer growth with metabolism, these clinical results could arguably be improved upon. In this Review, we discuss past and present efforts to overcome the somewhat limited clinical efficacy of PI3Kα pathway inhibitors, including optimization of inhibitor specificity, patient selection and biomarkers across cancer types, with a focus on breast cancer, as well as identification and abrogation of signalling-related and metabolic mechanisms of resistance, and interventions to improve management of prohibitive adverse events. We highlight the advantages and limitations of laboratory-based model systems used to study the PI3K pathway, and propose technologies and experimental inquiries to guide the future clinical deployment of PI3K pathway inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Neil Vasan
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
29
|
Kirchenwitz M, Stahnke S, Prettin S, Borowiak M, Menke L, Sieben C, Birchmeier C, Rottner K, Stradal TEB, Steffen A. SMER28 Attenuates PI3K/mTOR Signaling by Direct Inhibition of PI3K p110 Delta. Cells 2022; 11:1648. [PMID: 35626685 PMCID: PMC9140127 DOI: 10.3390/cells11101648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
SMER28 (Small molecule enhancer of Rapamycin 28) is an autophagy-inducing compound functioning by a hitherto unknown mechanism. Here, we confirm its autophagy-inducing effect by assessing classical autophagy-related parameters. Interestingly, we also discovered several additional effects of SMER28, including growth retardation and reduced G1 to S phase progression. Most strikingly, SMER28 treatment led to a complete arrest of receptor tyrosine kinase signaling, and, consequently, growth factor-induced cell scattering and dorsal ruffle formation. This coincided with a dramatic reduction in phosphorylation patterns of PI3K downstream effectors. Consistently, SMER28 directly inhibited PI3Kδ and to a lesser extent p110γ. The biological relevance of our observations was underscored by SMER28 interfering with InlB-mediated host cell entry of Listeria monocytogenes, which requires signaling through the prominent receptor tyrosine kinase c-Met. This effect was signaling-specific, since entry of unrelated, gram-negative Salmonella Typhimurium was not inhibited. Lastly, in B cell lymphoma cells, which predominantly depend on tonic signaling through PI3Kδ, apoptosis upon SMER28 treatment is profound in comparison to non-hematopoietic cells. This indicates SMER28 as a possible drug candidate for the treatment of diseases that derive from aberrant PI3Kδ activity.
Collapse
Affiliation(s)
- Marco Kirchenwitz
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Stephanie Stahnke
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
| | - Silvia Prettin
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
| | - Malgorzata Borowiak
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany; (M.B.); (C.B.)
| | - Laura Menke
- Nanoscale Infection Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
| | - Christian Sieben
- Nanoscale Infection Biology Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (L.M.); (C.S.)
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany; (M.B.); (C.B.)
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Theresia E. B. Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (M.K.); (S.S.); (S.P.); (K.R.)
| |
Collapse
|
30
|
Zhang C, Liu X, Wu H, Wang Y, Fan Y, Guo B, Bian X, Li X, Zhang J. Proteomic Response Revealed Signaling Pathways Involving in the Mechanism of Polymyxin B-Induced Melanogenesis. Microbiol Spectr 2022; 10:e0273021. [PMID: 35377227 PMCID: PMC9045165 DOI: 10.1128/spectrum.02730-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/05/2022] [Indexed: 12/03/2022] Open
Abstract
Polymyxin B is a last-line antibiotic for extensively resistant Gram-negative bacterial infection. Skin hyperpigmentation is a serious side effect induced by polymyxin B that severely compromises the psychological health and compliance of patients. The literature lacks mechanistic studies that explain how hyperpigmentation occurs, and this substantially hinders the development of intervention strategies and improved compliance. SK-MEL-2 cells were used for the polymyxin B-induced hyperpigmentation mechanism study. Melanin content and tyrosinase activity were measured after polymyxin B treatment. Tandem mass tag (TMT)-labeling quantitative proteomics was employed to investigate the response of SK-MEL-2 cells to polymyxin B treatment. Real-time quantitative PCR and Western blot were applied to validate the mRNA and protein levels of related genes and proteins. The melanin content and tyrosinase activity were significantly upregulated after polymyxin B treatment in SK-MEL-2 cells at 48 h and 72 h. Quantitative proteomics showed that 237 proteins were upregulated and 153 proteins were downregulated in the 48 h group, and 49 proteins were upregulated and 49 proteins were downregulated in the 72 h group. The differentially expressed proteins were involved in pathways such as lysosome, PI3K/Akt signaling pathway, and calcium signaling pathway. The upregulation of melanogenic enzymes and microphthalmia-associated transcription factor (MITF) was validated by qPCR and Western blot. Meanwhile, phosphorylation of PI3K, β-catenin, and cyclic-AMP response binding protein (CREB) in response to polymyxin B treatment was observed. The present study reveals the proteomic response of polymyxin B-induced melanogenesis in SK-MEL-2 cells for the first time. Signaling pathways, including melanin biosynthesis, PI3K/Akt, and calcium signaling pathways may be involved in the mechanism of melanogenesis. IMPORTANCE Polymyxin B-induced skin hyperpigmentation seriously affects the psychological health and compliance of patients. This study provides a proteomic clue to the mechanism at the cellular level for understanding polymyxin B-induced hyperpigmentation, contributing to a follow-up investigation of the corresponding PI3K/Akt signaling transduction pathway and calcium signaling pathway. The elucidation of its underlying mechanism is of great significance for patients' compliance improvement, intervention strategy, and new drug development.
Collapse
Affiliation(s)
- Chuhan Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hailan Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaxin Fan
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beining Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xingchen Bian
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People’s Republic of China, Shanghai, China
- National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Phase I Clinical Trial Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Jenkins ML, Ranga-prasad H, Parson MA, Harris NJ, Rathinaswamy MK, Burke JE. Oncogenic mutations of PIK3CAlead to increased membrane recruitment driven by reorientation of the ABD, p85 and C-terminus.. [DOI: 10.1101/2022.04.05.487205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractPIK3CAencoding the phosphoinositide 3-kinase (PI3K) p110α catalytic subunit is frequently mutated in cancer, with mutations occurring widely throughout the primary sequence. The full set of mechanisms underlying how PI3Ks are activated by all oncogenic mutations on membranes are unclear. Using a synergy of biochemical assays and hydrogen deuterium exchange mass spectrometry (HDX-MS), we reveal unique regulatory mechanisms underlying PI3K activation. Engagement of p110α on membranes leads to disengagement of the ABD of p110α from the catalytic core, and the C2 domain from the iSH2 domain of the p85 regulatory subunit. PI3K activation also requires reorientation of the p110α C-terminus, with mutations that alter the inhibited conformation of the C-terminus increasing membrane binding. Mutations at the C-terminus (M1043I/L, H1047R, G1049R, and N1068KLKR) activate p110α through distinct mechanisms, with this having important implications for mutant selective inhibitor development. This work reveals unique mechanisms underlying how PI3K is activated by oncogenic mutations, and explains how double mutants can synergistically increase PI3K activity.
Collapse
|
32
|
Valls PO, Esposito A. Signalling dynamics, cell decisions, and homeostatic control in health and disease. Curr Opin Cell Biol 2022; 75:102066. [PMID: 35245783 PMCID: PMC9097822 DOI: 10.1016/j.ceb.2022.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/13/2022]
Abstract
Cell signalling engenders cells with the capability to receive and process information from the intracellular and extracellular environments, trigger and execute biological responses, and communicate with each other. Ultimately, cell signalling is responsible for maintaining homeostasis at the cellular, tissue and systemic level. For this reason, cell signalling is a topic of intense research efforts aimed to elucidate how cells coordinate transitions between states in developing and adult organisms in physiological and pathological conditions. Here, we review current knowledge of how cell signalling operates at multiple spatial and temporal scales, focusing on how single-cell analytical techniques reveal mechanisms underpinning cell-to-cell variability, signalling plasticity, and collective cellular responses.
Collapse
Affiliation(s)
- Pablo Oriol Valls
- MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, United Kingdom
| | - Alessandro Esposito
- MRC Cancer Unit, University of Cambridge, Cambridge, CB2 0XZ, United Kingdom; Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, United Kingdom.
| |
Collapse
|
33
|
PI3K and AKT at the Interface of Signaling and Metabolism. Curr Top Microbiol Immunol 2022; 436:311-336. [DOI: 10.1007/978-3-031-06566-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
|
35
|
Zhang Y, He L, Chen X, Shentu P, Xu Y, Jiao J. Omega-3 polyunsaturated fatty acids promote SNAREs mediated GLUT4 vesicle docking and fusion. J Nutr Biochem 2021; 101:108912. [PMID: 34801692 DOI: 10.1016/j.jnutbio.2021.108912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 11/27/2022]
Abstract
Glucose homeostasis imbalance and insulin resistance (IR) are major contributors to the incidence of type 2 diabetes. Omega-3 polyunsaturated fatty acids (PUFAs) are key ingredients for maintaining cellular functions and improving insulin sensitivity. However, how omega-3 PUFAs modulate the dynamic process of glucose transport at the cellular level remains unclear. Here we unraveled eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may regulate the glucose transporter 4 (GLUT4) vesicle trafficking in both normal and IR adipocytes. Both omega-3 PUFAs significantly increase glucose consumption within a range of 10-32% in the basal state. Furthermore, both EPA (200 μM) and DHA (100 μM) may significantly promote the serine/threonine protein kinase (Akt) phosphorylation by 70% and 40% in the physiological state of adipocytes, respectively. Both omega-3 PUFAs significantly advanced the Akt phosphorylation in a dose-dependent way and showed a ∼2-fold increase at the dose of 200 μM in the IR pathological state. However, they could not up-regulate the expression of GLUT4 and insulin-regulated aminopeptidase protein. We further revealed that both omega-3 PUFAs dynamically promote insulin-stimulated GLUT4 vesicle translocation and soluble N-ethylmaleimide-sensitive factor attachment protein receptor mediated vesicle docking and fusion to the plasma membrane via specifically modulating the expression of vesicle-associated membrane protein 2. Understanding the mechanisms by which omega-3 PUFAs modulate cellular metabolism and IR in peripheral tissues may provide novel insights into the potential impact of omega-3 PUFAs on the metabolic function and the management of IR.
Collapse
Affiliation(s)
- Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lilin He
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoqian Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ping Shentu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yingke Xu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, China; Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
36
|
Vanhaesebroeck B, Perry MWD, Brown JR, André F, Okkenhaug K. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov 2021; 20:741-769. [PMID: 34127844 PMCID: PMC9297732 DOI: 10.1038/s41573-021-00209-1] [Citation(s) in RCA: 269] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 01/08/2023]
Abstract
Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval - the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addition to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clinical translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities.
Collapse
Affiliation(s)
| | - Matthew W D Perry
- Medicinal Chemistry, Research and Early Development, Respiratory & Immunology BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jennifer R Brown
- CLL Center, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Fabrice André
- Institut Gustave Roussy, INSERM U981, Université Paris Saclay, Paris, France
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Sipieter F, Cappe B, Leray A, De Schutter E, Bridelance J, Hulpiau P, Van Camp G, Declercq W, Héliot L, Vincent P, Vandenabeele P, Riquet FB. Characteristic ERK1/2 signaling dynamics distinguishes necroptosis from apoptosis. iScience 2021; 24:103074. [PMID: 34568795 PMCID: PMC8449238 DOI: 10.1016/j.isci.2021.103074] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/21/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
ERK1/2 involvement in cell death remains unclear, although many studies have demonstrated the importance of ERK1/2 dynamics in determining cellular responses. To untangle how ERK1/2 contributes to two cell death programs, we investigated ERK1/2 signaling dynamics during hFasL-induced apoptosis and TNF-induced necroptosis in L929 cells. We observed that ERK1/2 inhibition sensitizes cells to apoptosis while delaying necroptosis. By monitoring ERK1/2 activity by live-cell imaging using an improved ERK1/2 biosensor (EKAR4.0), we reported differential ERK1/2 signaling dynamics between cell survival, apoptosis, and necroptosis. We also decrypted a temporally shifted amplitude- and frequency-modulated (AM/FM) ERK1/2 activity profile in necroptosis versus apoptosis. ERK1/2 inhibition, which disrupted ERK1/2 signaling dynamics, prevented TNF and IL-6 gene expression increase during TNF-induced necroptosis. Using an inducible cell line for activated MLKL, the final executioner of necroptosis, we showed ERK1/2 and its distinctive necroptotic ERK1/2 activity dynamics to be positioned downstream of MLKL.
Collapse
Affiliation(s)
- François Sipieter
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium.,Université de Lille, Lille, France
| | - Benjamin Cappe
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium
| | - Aymeric Leray
- Laboratoire Interdisciplinaire Carnot De Bourgogne, UMR 6303 CNRS-Université de Bourgogne Franche-Comté, 21078 Dijon, France
| | - Elke De Schutter
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium.,Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, Edegem, 2650 Antwerp, Belgium
| | - Jolien Bridelance
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium
| | - Paco Hulpiau
- Data Mining and Modeling for Biomedicine (DaMBi), VIB Center for Inflammation Research, Ghent, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, Edegem, 2650 Antwerp, Belgium.,Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Wim Declercq
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium
| | - Laurent Héliot
- Team Biophotonique Cellulaire Fonctionnelle, Laboratoire de Physique des Lasers, Atomes et Molécules (PhLAM), CNRS UMR 8523, 59655 Villeneuve d'Ascq, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Neurobiology of Adaptative Processes, UMR8256, 75005 Paris, France
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium
| | - Franck B Riquet
- Molecular Signaling and Cell Death Unit, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Technologiepark 71, Zwijnaarde, 9052 Ghent, Belgium.,Université de Lille, Lille, France
| |
Collapse
|
38
|
Müller R, Kojic A, Citir M, Schultz C. Synthesis and Cellular Labeling of Multifunctional Phosphatidylinositol Bis- and Trisphosphate Derivatives. Angew Chem Int Ed Engl 2021; 60:19759-19765. [PMID: 34075669 PMCID: PMC8390440 DOI: 10.1002/anie.202103599] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/07/2021] [Indexed: 12/15/2022]
Abstract
We synthesized the first multifunctionalized phosphoinositide polyphosphate derivatives featuring a photo-removable protecting group ("cage"), a photo-crosslinkable diazirine group, and a terminal alkyne group useful for click chemistry. We demonstrate that the lipid derivatives readily enter cells. After photo-crosslinking, cell fixation and fluorescent tagging via click chemistry, we determined the intracellular location of the lipid derivatives before and after uncaging of the lipids. We find that there is rapid trafficking of PI(3,4)P2 and PI(3,4,5)P3 derivatives to the plasma membrane, opening the intriguing possibility that there is active transport of these lipids involved. We employed the photo-crosslinking and click chemistry functions to analyze the proteome of PI(3,4,5)P3 -binding proteins. From the latter, we validated by RNAi that the putative lipid binding proteins ATP11A and MPP6 are involved in the transport of PI(3,4,5)P3 to the plasma membrane.
Collapse
Affiliation(s)
- Rainer Müller
- European Molecular Biology Laboratory (EMBL)Cell Biology & Biophysics UnitMeyerhofstr. 169117HeidelbergGermany
| | - Ana Kojic
- European Molecular Biology Laboratory (EMBL)Cell Biology & Biophysics UnitMeyerhofstr. 169117HeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint Ph.D. Degree between EMBL and Heidelberg University69117HeidelbergGermany
- Oregon Health & Science UniversityDepartment of Chemical Physiology and Biochemistry3181 SW Sam Jackson Park RdPortlandOR97239-3098USA
| | - Mevlut Citir
- European Molecular Biology Laboratory (EMBL)Cell Biology & Biophysics UnitMeyerhofstr. 169117HeidelbergGermany
- Faculty of BiosciencesCollaboration for Joint Ph.D. Degree between EMBL and Heidelberg University69117HeidelbergGermany
| | - Carsten Schultz
- European Molecular Biology Laboratory (EMBL)Cell Biology & Biophysics UnitMeyerhofstr. 169117HeidelbergGermany
- Oregon Health & Science UniversityDepartment of Chemical Physiology and Biochemistry3181 SW Sam Jackson Park RdPortlandOR97239-3098USA
| |
Collapse
|
39
|
Müller R, Kojic A, Citir M, Schultz C. Synthesis and Cellular Labeling of Multifunctional Phosphatidylinositol Bis‐ and Trisphosphate Derivatives. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Rainer Müller
- European Molecular Biology Laboratory (EMBL) Cell Biology & Biophysics Unit Meyerhofstr. 1 69117 Heidelberg Germany
| | - Ana Kojic
- European Molecular Biology Laboratory (EMBL) Cell Biology & Biophysics Unit Meyerhofstr. 1 69117 Heidelberg Germany
- Faculty of Biosciences Collaboration for Joint Ph.D. Degree between EMBL and Heidelberg University 69117 Heidelberg Germany
- Oregon Health & Science University Department of Chemical Physiology and Biochemistry 3181 SW Sam Jackson Park Rd Portland OR 97239-3098 USA
| | - Mevlut Citir
- European Molecular Biology Laboratory (EMBL) Cell Biology & Biophysics Unit Meyerhofstr. 1 69117 Heidelberg Germany
- Faculty of Biosciences Collaboration for Joint Ph.D. Degree between EMBL and Heidelberg University 69117 Heidelberg Germany
| | - Carsten Schultz
- European Molecular Biology Laboratory (EMBL) Cell Biology & Biophysics Unit Meyerhofstr. 1 69117 Heidelberg Germany
- Oregon Health & Science University Department of Chemical Physiology and Biochemistry 3181 SW Sam Jackson Park Rd Portland OR 97239-3098 USA
| |
Collapse
|
40
|
Rathinaswamy MK, Fleming KD, Dalwadi U, Pardon E, Harris NJ, Yip CK, Steyaert J, Burke JE. HDX-MS-optimized approach to characterize nanobodies as tools for biochemical and structural studies of class IB phosphoinositide 3-kinases. Structure 2021; 29:1371-1381.e6. [PMID: 34348129 DOI: 10.1016/j.str.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/07/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
There is considerable interest in developing antibodies as modulators of signaling pathways. One of the most important signaling pathways in higher eukaryotes is the phosphoinositide 3-kinase (PI3K) pathway, which plays fundamental roles in growth, metabolism, and immunity. The class IB PI3K, PI3Kγ, is a heterodimeric complex composed of a catalytic p110γ subunit bound to a p101 or p84 regulatory subunit. PI3Kγ is a critical component in multiple immune signaling processes and is dependent on activation by Ras and G protein-coupled receptors (GPCRs) to mediate its cellular roles. Here we describe the rapid and efficient characterization of multiple PI3Kγ binding single-chain camelid nanobodies using hydrogen-deuterium exchange (HDX) mass spectrometry (MS) for structural and biochemical studies. We identify nanobodies that stimulated lipid kinase activity, block Ras activation, and specifically inhibited p101-mediated GPCR activation. Overall, our work reveals insight into PI3Kγ regulation and identifies sites that may be exploited for therapeutic development.
Collapse
Affiliation(s)
- Manoj K Rathinaswamy
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Kaelin D Fleming
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Udit Dalwadi
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Noah J Harris
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Calvin K Yip
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium; VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada; Life Sciences Institute, Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
41
|
Insulin-mediated immune dysfunction in the development of preeclampsia. J Mol Med (Berl) 2021; 99:889-897. [PMID: 33768298 DOI: 10.1007/s00109-021-02068-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 02/08/2023]
Abstract
Epidemiological observations implicate insulin resistance as a predisposing factor in the development of preeclampsia (PE). It is also well established that PE manifests in the context of a dysregulated immune response at the maternal-foetal interface, though all the underlying drivers of such immune dysregulation remains to be accounted for. Although it has long been known that various immune cells express insulin receptors following immune activation, it is only recently that insulin signalling has been shown to play a key role in immune cell differentiation, survival and effector function through its canonical activation of the PI3K/Akt/mTOR pathway. Here we argue that hyperinsulinemia, manifesting either from insulin resistance or from intensive insulin therapy, likely plays a direct role in driving immune cell dysfunction which plays a central role in the development of PE. This line of reasoning also explains the superior results of insulin-sparing interventions compared to intensive insulin therapy as monotherapy.
Collapse
|
42
|
van Niekerk G, van der Merwe M, Engelbrecht AM. Diabetes and susceptibility to infections: Implication for COVID-19. Immunology 2021; 164:467-475. [PMID: 34115881 PMCID: PMC8446942 DOI: 10.1111/imm.13383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/20/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022] Open
Abstract
A number of mechanisms have been proposed to explain the well‐established link between diabetic status and an increased susceptibility to infection. Notably, diabetes has been shown to be one of the strongest factors influencing healthcare outcome in COVID‐19 infections. Though it has long been noted that lymphocytes upregulate insulin receptors following immune activation, until recently, this observation has received little attention. Here, we point out key findings implicating dysregulated insulin signalling in immune cells as a possible contributing factor in the immune pathology associated with diabetes. Mechanistically, insulin, by activating the PI3K/Akt/mTOR pathway, regulates various aspects of both myeloid cells and lymphocytes, such as cell survival, metabolic reprogramming and the polarization and differentiation of immune cells. PI3K signalling is also supressed by immune checkpoint proteins, suggesting that insulin signalling may antagonize peripheral tolerance. Remarkably, it has also recently been shown that, following insulin binding, the insulin receptor translocates to the nucleus where it plays a key role in regulating the transcription of various immune‐related genes, including pathways involved in viral infections. Taken together, these observations suggest that dysregulated insulin signalling may directly contribute to a defective immune response during COVID‐19 infections.
Collapse
Affiliation(s)
- Gustav van Niekerk
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Michelle van der Merwe
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
43
|
Lampson BL, Brown JR. The Evolving Use of Phosphatidylinositol 3-Kinase Inhibitors for the Treatment of Chronic Lymphocytic Leukemia. Hematol Oncol Clin North Am 2021; 35:807-826. [PMID: 34174987 DOI: 10.1016/j.hoc.2021.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
B cells express 4 phosphatidylinositol 3-kinase (PI3K) isoforms and have a dependence on p110δ for survival. The design of isoform-selective inhibitors is possible, and pharmacologic inhibition of p110δ is toxic to neoplastic chronic lymphocytic leukemia (CLL) cells for both cell-intrinsic and cell-extrinsic reasons. Idelalisib is a first-in-class p110δ inhibitor that exhibits efficacy for the treatment of relapsed CLL irrespective of adverse prognostic features. Duvelisib is a p110γ/δ inhibitor with a similar efficacy and safety profile to idelalisib. Recent data indicate that umbralisib, a p110δ/CK-1ε dual inhibitor, is safe and effective when administered to patients with CLL.
Collapse
Affiliation(s)
- Benjamin L Lampson
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Jennifer R Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, CLL Center, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
44
|
Rathinaswamy MK, Gaieb Z, Fleming KD, Borsari C, Harris NJ, Moeller BE, Wymann MP, Amaro RE, Burke JE. Disease-related mutations in PI3Kγ disrupt regulatory C-terminal dynamics and reveal a path to selective inhibitors. eLife 2021; 10:e64691. [PMID: 33661099 PMCID: PMC7955810 DOI: 10.7554/elife.64691] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Class I Phosphoinositide 3-kinases (PI3Ks) are master regulators of cellular functions, with the class IB PI3K catalytic subunit (p110γ) playing key roles in immune signalling. p110γ is a key factor in inflammatory diseases and has been identified as a therapeutic target for cancers due to its immunomodulatory role. Using a combined biochemical/biophysical approach, we have revealed insight into regulation of kinase activity, specifically defining how immunodeficiency and oncogenic mutations of R1021 in the C-terminus can inactivate or activate enzyme activity. Screening of inhibitors using HDX-MS revealed that activation loop-binding inhibitors induce allosteric conformational changes that mimic those in the R1021C mutant. Structural analysis of advanced PI3K inhibitors in clinical development revealed novel binding pockets that can be exploited for further therapeutic development. Overall, this work provides unique insights into regulatory mechanisms that control PI3Kγ kinase activity and shows a framework for the design of PI3K isoform and mutant selective inhibitors.
Collapse
Affiliation(s)
- Manoj K Rathinaswamy
- Department of Biochemistry and Microbiology, University of VictoriaVictoriaCanada
| | - Zied Gaieb
- Department of Chemistry and Biochemistry, University of California San DiegoSan DiegoUnited States
| | - Kaelin D Fleming
- Department of Biochemistry and Microbiology, University of VictoriaVictoriaCanada
| | - Chiara Borsari
- University of Basel, Department of BiomedicineBaselSwitzerland
| | - Noah J Harris
- Department of Biochemistry and Microbiology, University of VictoriaVictoriaCanada
| | - Brandon E Moeller
- Department of Biochemistry and Microbiology, University of VictoriaVictoriaCanada
| | | | - Rommie E Amaro
- Department of Chemistry and Biochemistry, University of California San DiegoSan DiegoUnited States
| | - John E Burke
- Department of Biochemistry and Microbiology, University of VictoriaVictoriaCanada
- Department of Biochemistry and Molecular Biology, The University of British ColumbiaVancouverCanada
| |
Collapse
|
45
|
The G-Protein Rab5A Activates VPS34 Complex II, a Class III PI3K, by a Dual Regulatory Mechanism. Biophys J 2020; 119:2205-2218. [PMID: 33137306 DOI: 10.1016/j.bpj.2020.10.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/04/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
VPS34 complex II (VPS34CII) is a 386-kDa assembly of the lipid kinase subunit VPS34 and three regulatory subunits that altogether function as a prototypical class III phosphatidylinositol-3-kinase (PI3K). When the active VPS34CII complex is docked to the cytoplasmic surface of endosomal membranes, it phosphorylates its substrate lipid (phosphatidylinositol, PI) to generate the essential signaling lipid phosphatidylinositol-3-phosphate (PI3P). In turn, PI3P recruits an array of signaling proteins containing PI3P-specific targeting domains (including FYVE, PX, and PROPPINS) to the membrane surface, where they initiate key cell processes. In endocytosis and early endosome development, net VPS34CII-catalyzed PI3P production is greatly amplified by Rab5A, a small G protein of the Ras GTPase superfamily. Moreover, VPS34CII and Rab5A are each strongly linked to multiple human diseases. Thus, a molecular understanding of the mechanism by which Rab5A activates lipid kinase activity will have broad impacts in both signaling biology and medicine. Two general mechanistic models have been proposed for small G protein activation of PI3K lipid kinases. 1) In the membrane recruitment mechanism, G protein association increases the density of active kinase on the membrane. And 2) in the allosteric activation mechanism, G protein allosterically triggers an increase in the specific activity (turnover rate) of the membrane-bound kinase molecule. This study employs an in vitro single-molecule approach to elucidate the mechanism of GTP-Rab5A-associated VPS34CII kinase activation in a reconstituted GTP-Rab5A-VPS34CII-PI3P-PX signaling pathway on a target membrane surface. The findings reveal that both membrane recruitment and allosteric mechanisms make important contributions to the large increase in VPS34CII kinase activity and PI3P production triggered by membrane-anchored GTP-Rab5A. Notably, under near-physiological conditions in the absence of other activators, membrane-anchored GTP-Rab5A provides strong, virtually binary on-off switching and is required for VPS34CII membrane binding and PI3P production.
Collapse
|
46
|
Perdomo J, Quintana C, González I, Hernández I, Rubio S, Loro JF, Reiter RJ, Estévez F, Quintana J. Melatonin Induces Melanogenesis in Human SK-MEL-1 Melanoma Cells Involving Glycogen Synthase Kinase-3 and Reactive Oxygen Species. Int J Mol Sci 2020; 21:ijms21144970. [PMID: 32674468 PMCID: PMC7404125 DOI: 10.3390/ijms21144970] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/12/2022] Open
Abstract
Melatonin is present in all living organisms where it displays a diversity of physiological functions. Attenuation of melanogenesis by melatonin has been reported in some mammals and also in rodent melanoma cells. However, melatonin may also stimulate melanogenesis in human melanoma cells through mechanisms that have not yet been revealed. Using the human melanoma cells SK-MEL-1 as a model, an increase in both tyrosinase activity and melanin was already observed at 24 h after melatonin treatment with maximal levels of both being detected at 72 h. This effect was associated with the induction in the expression of the enzymes involved in the synthesis of melanin. In this scenario, glycogen synthase kinase-3β seems to play a significant function since melatonin decreased its phosphorylation and preincubation with specific inhibitors of this protein kinase (lithium or BIO) reduced the expression and activity of tyrosinase. Blocking of PI3K/AKT pathway stimulated melanogenesis and the effect was suppressed by the inhibitors of glycogen synthase kinase-3β. Although melatonin is a recognized antioxidant, we found that it stimulates reactive oxygen species generation in SK-MEL-1 cells. These chemical species seem to be an important signal in activating the melanogenic process since the antioxidants N-acetyl-l-cysteine and glutathione decreased both the level and activity of tyrosinase stimulated by melatonin. Our results support the view that regulation of melanogenesis involves a cross-talk between several signaling pathways.
Collapse
Affiliation(s)
- Juan Perdomo
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - Carlos Quintana
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - Ignacio González
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - Inmaculada Hernández
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - Sara Rubio
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - Juan F. Loro
- Departamento de Ciencias Clínicas, Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain;
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science at San Antonio, San Antonio, TX 78229, USA;
| | - Francisco Estévez
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
| | - José Quintana
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de las Palmas de Gran Canaria, 35016 Las Palmas, Spain; (J.P.); (C.Q.); (I.G.); (I.H.); (S.R.); (F.E.)
- Correspondence: ; Tel.: +34-928458792
| |
Collapse
|
47
|
Madsen RR. PI3K in stemness regulation: from development to cancer. Biochem Soc Trans 2020; 48:301-315. [PMID: 32010943 PMCID: PMC7054754 DOI: 10.1042/bst20190778] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
Abstract
The PI3K/AKT pathway is a key target in oncology where most efforts are focussed on phenotypes such as cell proliferation and survival. Comparatively, little attention has been paid to PI3K in stemness regulation, despite the emerging link between acquisition of stem cell-like features and therapeutic failure in cancer. The aim of this review is to summarise current known and unknowns of PI3K-dependent stemness regulation, by integrating knowledge from the fields of developmental, signalling and cancer biology. Particular attention is given to the role of the PI3K pathway in pluripotent stem cells (PSCs) and the emerging parallels to dedifferentiated cancer cells with stem cell-like features. Compelling evidence suggests that PI3K/AKT signalling forms part of a 'core molecular stemness programme' in both mouse and human PSCs. In cancer, the oncogenic PIK3CAH1047R variant causes constitutive activation of the PI3K pathway and has recently been linked to increased stemness in a dose-dependent manner, similar to observations in mouse PSCs with heterozygous versus homozygous Pten loss. There is also evidence that the stemness phenotype may become 'locked' and thus independent of the original PI3K activation, posing limitations for the success of PI3K monotherapy in cancer. Ongoing therapeutic developments for PI3K-associated cancers may therefore benefit from a better understanding of the pathway's two-layered and highly context-dependent regulation of cell growth versus stemness.
Collapse
Affiliation(s)
- Ralitsa R. Madsen
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E 6DD, U.K
| |
Collapse
|
48
|
Giguère V. DNA-PK, Nuclear mTOR, and the Androgen Pathway in Prostate Cancer. Trends Cancer 2020; 6:337-347. [PMID: 32209447 DOI: 10.1016/j.trecan.2020.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
Androgen and its receptor (AR) are major drivers of prostate cancer (PCa), a leading cause of mortality in aging men. Thus, understanding the numerous mechanisms by which AR can promote the growth and proliferation of PCa cells and enable their escape from hormone-dependent therapies, eventually leading to metastasis and death of the patient, is essential to discover alternative therapeutic approaches. Recently, two structurally related members of the phosphatidylinositol 3-kinase-like protein kinase (PIKK) family, DNA-dependent protein kinase (DNA-PK) and mammalian target of rapamycin (mTOR), were shown to have a direct role in modulating AR activity on chromatin of PCa cells. In this review, the common features of DNA-PK and mTOR and the similarities in their noncanonical roles as transcription coregulators of the AR are highlighted. An outlook on how these findings could be translated into new approaches to manage and treat PCa is provided.
Collapse
Affiliation(s)
- Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
49
|
Mátyási B, Farkas Z, Kopper L, Sebestyén A, Boissan M, Mehta A, Takács-Vellai K. The Function of NM23-H1/NME1 and Its Homologs in Major Processes Linked to Metastasis. Pathol Oncol Res 2020; 26:49-61. [PMID: 31993913 PMCID: PMC7109179 DOI: 10.1007/s12253-020-00797-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Metastasis suppressor genes (MSGs) inhibit different biological processes during metastatic progression without globally influencing development of the primary tumor. The first MSG, NM23 (non-metastatic clone 23, isoform H1) or now called NME1 (stands for non-metastatic) was identified some decades ago. Since then, ten human NM23 paralogs forming two groups have been discovered. Group I NM23 genes encode enzymes with evolutionarily highly conserved nucleoside diphosphate kinase (NDPK) activity. In this review we summarize how results from NDPKs in model organisms converged on human NM23 studies. Next, we examine the role of NM23-H1 and its homologs within the metastatic cascade, e.g. cell migration and invasion, proliferation and apoptosis. NM23-H1 homologs are well known inhibitors of cell migration. Drosophila studies revealed that AWD, the fly counterpart of NM23-H1 is a negative regulator of cell motility by modulating endocytosis of chemotactic receptors on the surface of migrating cells in cooperation with Shibire/Dynamin; this mechanism has been recently confirmed by human studies. NM23-H1 inhibits proliferation of tumor cells by phosphorylating the MAPK scaffold, kinase suppressor of Ras (KSR), resulting in suppression of MAPK signalling. This mechanism was also observed with the C. elegans homolog, NDK-1, albeit with an inverse effect on MAPK activation. Both NM23-H1 and NDK-1 promote apoptotic cell death. In addition, NDK-1, NM23-H1 and their mouse counterpart NM23-M1 were shown to promote phagocytosis in an evolutionarily conserved manner. In summary, inhibition of cell migration and proliferation, alongside actions in apoptosis and phagocytosis are all mechanisms through which NM23-H1 acts against metastatic progression.
Collapse
Affiliation(s)
- Barbara Mátyási
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117, Budapest, Hungary
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117, Budapest, Hungary
| | - László Kopper
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1st, Budapest, Hungary
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1st, Budapest, Hungary
| | - Mathieu Boissan
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
- Service de Biochimie et Hormonologie, AP- HP, Hôpital Tenon, Paris, France
| | - Anil Mehta
- Division of Medical Sciences, Centre for CVS and Lung Biology, Ninewells Hospital Medical School, DD19SY, Dundee, UK
| | - Krisztina Takács-Vellai
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter stny. 1/C, H-1117, Budapest, Hungary.
| |
Collapse
|