1
|
Toscano E, Cimmino E, Boccia A, Sepe L, Paolella G. Cell populations simulated in silico within SimulCell accurately reproduce the behaviour of experimental cell cultures. NPJ Syst Biol Appl 2025; 11:48. [PMID: 40379622 DOI: 10.1038/s41540-025-00518-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/08/2025] [Indexed: 05/19/2025] Open
Abstract
In silico simulations are used to understand cell behaviour by means of different approaches and tools, which range from reproducing average population trends to building lattice-based models to, more recently, creating populations of individual cell agents whose mass, volume and morphology behave according to more or less precise rules and models. In this work, a new agent-based simulator, SimulCell, was conceived, developed and used to predict the behaviour of eukaryotic cell cultures while growing attached to a flat surface. The system, starting from time-lapse microscopy experiments, uses growth, proliferation and migration models to create synthetic populations closely resembling original cultures. Support for cell-cell and cell-environment interaction makes cell agents able to react to changes in medium composition and other events, such as physical damage or chemical modifications occurring in the culture plate. The simulator is accessible through a web application and generates data that can be shown as tables and graphs or exported for further analyses.
Collapse
Affiliation(s)
- Elvira Toscano
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - Elena Cimmino
- Department of Molecular Medicine and Medical Biotechnology, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - Angelo Boccia
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy
| | - Leandra Sepe
- Department of Molecular Medicine and Medical Biotechnology, Università Degli Studi di Napoli "Federico II", Naples, Italy
| | - Giovanni Paolella
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnology, Università Degli Studi di Napoli "Federico II", Naples, Italy.
| |
Collapse
|
2
|
Murley A, Popovici AC, Hu XS, Lund A, Wickham K, Durieux J, Joe L, Koronyo E, Zhang H, Genuth NR, Dillin A. Quiescent cell re-entry is limited by macroautophagy-induced lysosomal damage. Cell 2025; 188:2670-2686.e14. [PMID: 40203825 DOI: 10.1016/j.cell.2025.03.009] [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: 06/26/2024] [Revised: 01/14/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
To maintain tissue homeostasis, many cells reside in a quiescent state until prompted to divide. The reactivation of quiescent cells is perturbed with aging and may underlie declining tissue homeostasis and resiliency. The unfolded protein response regulators IRE-1 and XBP-1 are required for the reactivation of quiescent cells in developmentally L1-arrested C. elegans. Utilizing a forward genetic screen in C. elegans, we discovered that macroautophagy targets protein aggregates to lysosomes in quiescent cells, leading to lysosome damage. Genetic inhibition of macroautophagy and stimulation of lysosomes via the overexpression of HLH-30 (TFEB/TFE3) synergistically reduces lysosome damage. Damaged lysosomes require IRE-1/XBP-1 for their repair following prolonged L1 arrest. Protein aggregates are also targeted to lysosomes by macroautophagy in quiescent cultured mammalian cells and are associated with lysosome damage. Thus, lysosome damage is a hallmark of quiescent cells, and limiting lysosome damage by restraining macroautophagy can stimulate their reactivation.
Collapse
Affiliation(s)
- Andrew Murley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ann Catherine Popovici
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Xiwen Sophie Hu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Anina Lund
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kevin Wickham
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jenni Durieux
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Larry Joe
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Etai Koronyo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Hanlin Zhang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Naomi R Genuth
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Andrew Dillin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
3
|
Diaba F, Morán MDC, Teixidó E. From In Vitro Cytotoxicity to In Vivo Zebrafish Assays: A Study on 3,3-Dichloro β-, γ- and δ-Lactams and Their Biological Activity Profiles. Pharmaceuticals (Basel) 2025; 18:488. [PMID: 40283925 PMCID: PMC12030062 DOI: 10.3390/ph18040488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Anticancer research is a constantly evolving field due to cancer's complexity and adaptability. This study aims to evaluate the hemolytic behavior and cytotoxic properties of ten 3,3-dichlorolactams against A431 tumor and 3T3 fibroblast cells, with a particular focus on their selective toxicity. Methods: To achieve this, we assessed the hemocompatibility and cytotoxic effects of the lactams, determining their impact on cell viability through MTT and NRU assays. Additionally, AO/EtBr double staining was used to confirm apoptosis as a mechanism of cell death. To complement the in vitro findings, in vivo experiments were conducted using Zebrafish embryos to evaluate acute, developmental, and neurotoxic effects. Results: The results demonstrated that all lactams were hemocompatible, with the cytotoxicity influenced mainly by their structure and the tested concentration. β-Lactam 1 was the most efficient in inducing selective toxicity against A431 cells, showing the lowest IC50 values (71 μg/mL and 210 μg/mL (MTT) and 35 μg/mL and >250 μg/mL (NRU) for A431 and 3T3 cell lines, respectively), with SI values close to 3 and >7. Moreover, cell death induction through apoptosis was confirmed by AO/EtBr double staining. Finally, despite its lower acute toxicity compared to other anticancer agents, the in vivo experiments revealed that 1 induced developmental toxicity and neurotoxic effects in Zebrafish embryos at concentrations lower than those affecting A431 cancer cells. Conclusions: the study highlights the potential of β-lactam derivatives as promising anticancer agents while emphasizing the need for comprehensive safety assessments. Future research should further explore structural modifications to enhance efficacy and specificity while minimizing adverse effects.
Collapse
Affiliation(s)
- Faïza Diaba
- Laboratori de Química Orgànica, Facultat de Farmàcia i Ciències de l’Alimentació, IBUB, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - María del Carmen Morán
- Departament de Bioquímica i Fisiologia-Secció de Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentaciò, Universitat de Barcelona, Avda. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
| | - Elisabet Teixidó
- GRET-Unitat de Toxicologia, Departamento de Farmacología, Toxicología y Química Terapéutica, Facultat de Farmàcia i Ciències de l’Alimentació, INSA, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
| |
Collapse
|
4
|
Shameem M, Olson SL, Marron Fernandez de Velasco E, Kumar A, Singh BN. Cardiac Fibroblasts: Helping or Hurting. Genes (Basel) 2025; 16:381. [PMID: 40282342 PMCID: PMC12026832 DOI: 10.3390/genes16040381] [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: 02/28/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Cardiac fibroblasts (CFs) are the essential cell type for heart morphogenesis and homeostasis. In addition to maintaining the structural integrity of the heart tissue, muscle fibroblasts are involved in complex signaling cascades that regulate cardiomyocyte proliferation, migration, and maturation. While CFs serve as the primary source of extracellular matrix proteins (ECM), tissue repair, and paracrine signaling, they are also responsible for adverse pathological changes associated with cardiovascular disease. Following activation, fibroblasts produce excessive ECM components that ultimately lead to fibrosis and cardiac dysfunction. Decades of research have led to a much deeper understanding of the role of CFs in cardiogenesis. Recent studies using the single-cell genomic approach have focused on advancing the role of CFs in cellular interactions, and the mechanistic implications involved during cardiovascular development and disease. Arguably, the unique role of fibroblasts in development, tissue repair, and disease progression categorizes them into the friend or foe category. This brief review summarizes the current understanding of cardiac fibroblast biology and discusses the key findings in the context of development and pathophysiological conditions.
Collapse
Affiliation(s)
- Mohammad Shameem
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Shelby L. Olson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - Akhilesh Kumar
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bhairab N. Singh
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA;
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Poirier A, Utecht T, Villot R, Gélinas Y, Mouchiroud M, Kordahi M, Kolnohuz A, Pasteur C, Roy J, Beaulieu MJ, Orain M, Samson N, Blanchet MR, Joubert P, Laplante M. ZNF768 loss amplifies p53 action and reduces lung tumorigenesis in mice. Oncogene 2025:10.1038/s41388-025-03352-w. [PMID: 40133474 DOI: 10.1038/s41388-025-03352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Cell proliferation is a fundamental process required for organismal development, growth, and maintenance. Failure to control this process leads to several diseases, including cancer. Zinc finger protein 768 (ZNF768) is an emerging transcription factor that plays key roles in driving proliferation. In addition to controlling a gene network supporting cell division, ZNF768 physically interacts and inhibits the activity of the tumor suppressor p53. Although the importance of ZNF768 in promoting cell proliferation has been well demonstrated in vitro, the physiological and pathological roles of ZNF768 in vivo are still unknown. Here, we report the generation and characterization of a ZNF768 null mouse model. ZNF768 null mice are viable but show a growth defect early in life. Mouse embryonic fibroblasts (MEFs) isolated from ZNF768 null embryos exhibit higher p53 levels, premature senescence, and higher sensitivity to genotoxic stress. In line with these findings, ZNF768 null mice showed increased radiosensitivity. This effect was associated not only with higher expression of a subset of p53 target genes, but also with alterations in genes regulating transmembrane receptor signaling, cell adhesion, and growth. Because ZNF768 levels are elevated in tumors, we tested the impact of ZNF768 loss on cancer development in mice. Here, we show that ZNF768 deletion was sufficient to repress lung tumor development in a KRASG12D-induced cancer mouse model. Overall, our findings establish ZNF768 as an important protein controlling cell proliferation that could potentially be targeted to reduce tumorigenesis.
Collapse
Affiliation(s)
- Audrey Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Timon Utecht
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Romain Villot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yves Gélinas
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Manal Kordahi
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Alona Kolnohuz
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
| | - Coline Pasteur
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Joanny Roy
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Josée Beaulieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Michèle Orain
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Nolwenn Samson
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Philippe Joubert
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada.
- Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, QC, Canada.
- Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
6
|
Srinivasan S, Ho HYH. An Efficient Method for Immortalizing Mouse Embryonic Fibroblasts by CRISPR-mediated Deletion of the Tp53 Gene. Bio Protoc 2025; 15:e5159. [PMID: 39872719 PMCID: PMC11769752 DOI: 10.21769/bioprotoc.5159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Mouse embryonic fibroblasts (MEFs) derived from genetically modified mice are a valuable resource for studying gene function and regulation. The MEF system can also be combined with rescue studies to characterize the function of mutant genes/proteins, such as disease-causing variants. However, primary MEFs undergo senescence soon after isolation and passaging, making long-term genetic manipulations difficult. Previously described methods for MEF immortalization are often inconsistent or alter the physiological properties of the cells. Here, we describe an optimized method that overcomes these limitations. By using electroporation to deliver CRISPR constructs that target the Tp53 gene, the method reliably generates immortalized MEFs (iMEFs) within three weeks. Importantly, iMEFs closely resemble the parent cell populations, and individual iMEFs can be cloned and expanded for subsequent genetic manipulation and characterization. We envision that this protocol can be adopted broadly to immortalize other mouse primary cell types. Key features • CRISPR-based knockout of the Tp53 gene enables efficient immortalization of mouse embryonic fibroblasts (MEFs) in under three weeks. • Immortalization requires a Neon electroporator or a comparable system to transfect cells with the Tp53 CRISPR constructs.
Collapse
Affiliation(s)
- Srisathya Srinivasan
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA, USA
| |
Collapse
|
7
|
Haj M, Frey Y, Levon A, Maliah A, Ben-Yishay T, Slutsky R, Smoom R, Tzfati Y, Ben-David U, Levy C, Elkon R, Ziv Y, Shiloh Y. The cGAS-STING, p38 MAPK, and p53 pathways link genome instability to accelerated cellular senescence in ATM-deficient murine lung fibroblasts. Proc Natl Acad Sci U S A 2025; 122:e2419196122. [PMID: 39772747 PMCID: PMC11745328 DOI: 10.1073/pnas.2419196122] [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: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Ataxia-telangiectasia (A-T) is a pleiotropic genome instability syndrome resulting from the loss of the homeostatic protein kinase ATM. The complex phenotype of A-T includes progressive cerebellar degeneration, immunodeficiency, gonadal atrophy, interstitial lung disease, cancer predisposition, endocrine abnormalities, chromosomal instability, radiosensitivity, and segmental premature aging. Cultured skin fibroblasts from A-T patients exhibit premature senescence, highlighting the association between genome instability, cellular senescence, and aging. We found that lung fibroblasts derived from ATM-deficient mice provide a versatile experimental system to explore the mechanisms driving the premature senescence of primary fibroblasts lacking ATM. Atm-/- fibroblasts failed to proliferate under ambient oxygen conditions (21%). Although they initially proliferated under physiological oxygen levels (3%), they rapidly entered senescence. In contrast, wild-type (WT) lung fibroblasts did not senesce under 3% oxygen and eventually underwent immortalization and neoplastic transformation. However, rapid senescence could be induced in WT cells either by Atm gene ablation or persistent chemical inhibition of ATM kinase activity, with senescence induced by ATM inhibition being reversible upon inhibitor removal. Moreover, the concomitant loss of ATM and p53 led to senescence evasion, vigorous growth, rampant genome instability, and subsequent immortalization and transformation. Our findings reveal that the rapid senescence of Atm-/- lung fibroblasts is driven by the collaborative action of the cGAS-STING, p38 MAPK, and p53 pathways in response to persistent DNA damage, ultimately leading to the induction of interferon-α1 and downstream interferon-stimulated genes. We propose that accelerated cellular senescence may exacerbate specific A-T symptoms, particularly contributing to the progressive, life-threatening interstitial lung disease often observed in A-T patients during adulthood.
Collapse
Affiliation(s)
- Majd Haj
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Yann Frey
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Amit Levon
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Avishai Maliah
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Tal Ben-Yishay
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Rachel Slutsky
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Riham Smoom
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem9190501, Israel
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem9190501, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Carmit Levy
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Yael Ziv
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Yosef Shiloh
- Department of Human Molecular Genetics and Biochemistry, Faculty of Health & Medical Sciences, Tel Aviv University, Tel Aviv69978, Israel
| |
Collapse
|
8
|
Mann JP, Tábara LC, Patel S, Pushpa P, Alvarez-Guaita A, Dong L, Haider A, Lim K, Tandon P, Scurria F, Minchin JEN, O’Rahilly S. S, Fazakerley DJ, Prudent J, Semple RK, Savage DB. Loss of Mfn1 but not Mfn2 enhances adipogenesis. PLoS One 2024; 19:e0306243. [PMID: 39739772 PMCID: PMC11687706 DOI: 10.1371/journal.pone.0306243] [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: 05/26/2023] [Accepted: 06/13/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE A biallelic missense mutation in mitofusin 2 (MFN2) causes multiple symmetric lipomatosis and partial lipodystrophy, implicating disruption of mitochondrial fusion or interaction with other organelles in adipocyte differentiation, growth and/or survival. In this study, we aimed to document the impact of loss of mitofusin 1 (Mfn1) or 2 (Mfn2) on adipogenesis in cultured cells. METHODS We characterised adipocyte differentiation of wildtype (WT), Mfn1-/- and Mfn2-/- mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes in which Mfn1 or 2 levels were reduced using siRNA. RESULTS Mfn1-/- MEFs displayed striking fragmentation of the mitochondrial network, with surprisingly enhanced propensity to differentiate into adipocytes, as assessed by lipid accumulation, expression of adipocyte markers (Plin1, Fabp4, Glut4, Adipoq), and insulin-stimulated glucose uptake. RNA sequencing revealed a corresponding pro-adipogenic transcriptional profile including Pparg upregulation. Mfn2-/- MEFs also had a disrupted mitochondrial morphology, but in contrast to Mfn1-/- MEFs they showed reduced expression of adipocyte markers. Mfn1 and Mfn2 siRNA mediated knockdown studies in 3T3-L1 adipocytes generally replicated these findings. CONCLUSIONS Loss of Mfn1 but not Mfn2 in cultured pre-adipocyte models is pro-adipogenic. This suggests distinct, non-redundant roles for the two mitofusin orthologues in adipocyte differentiation.
Collapse
Affiliation(s)
- Jake P. Mann
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Luis Carlos Tábara
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Satish Patel
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Pushpa Pushpa
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Anna Alvarez-Guaita
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Liang Dong
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Afreen Haider
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Koini Lim
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Panna Tandon
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Fabio Scurria
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - James E. N. Minchin
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen O’Rahilly S.
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Daniel J. Fazakerley
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Julien Prudent
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Robert K. Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - David B. Savage
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Franks NE, Allen BL. Hedgehog-dependent and hedgehog-independent roles for growth arrest specific 1 in mammalian kidney morphogenesis. Development 2024; 151:dev203012. [PMID: 39629522 PMCID: PMC11795293 DOI: 10.1242/dev.203012] [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: 05/07/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024]
Abstract
Growth arrest specific 1 (GAS1) is a key regulator of mammalian embryogenesis, best known for its role in hedgehog (HH) signaling, but with additional described roles in the FGF, RET, and NOTCH pathways. Previous work indicated a later role for GAS1 in kidney development through FGF pathway modulation. Here, we demonstrate that GAS1 is essential for both mesonephrogenesis and metanephrogenesis - most notably, Gas1 deletion in mice results in renal agenesis in a genetic background-dependent fashion. Mechanistically, GAS1 promotes mesonephrogenesis in a HH-dependent fashion, performing a unique co-receptor function, while promoting metanephrogenesis in a HH-independent fashion, acting as a putative secreted RET co-receptor. Our data indicate that Gas1 deletion leads to renal agenesis through a transient reduction in metanephric mesenchyme proliferation - a phenotype that can be rescued by exogenous RET pathway stimulation. Overall, this study indicates that GAS1 contributes to early kidney development through the integration of multiple different signaling pathways.
Collapse
Affiliation(s)
- Nicole E. Franks
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Benjamin L. Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Nakajima K, Nishizawa H, Chen G, Tsuge S, Yamanaka M, Kiyohara M, Irikura R, Matsumoto M, Tanaka K, Narikawa R, Igarashi K. Intracellular biliverdin dynamics during ferroptosis. J Biochem 2024; 176:472-483. [PMID: 39340324 PMCID: PMC11638335 DOI: 10.1093/jb/mvae067] [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: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Ferroptosis is a cell death mechanism mediated by iron-dependent lipid peroxidation. Although ferroptosis has garnered attention as a cancer-suppressing mechanism, there are still limited markers available for identifying ferroptotic cells or assessing their sensitivity to ferroptosis. The study focused on biliverdin, an endogenous reducing substance in cells, and examined the dynamics of intracellular biliverdin during ferroptosis using a biliverdin-binding cyanobacteriochrome. It was found that intracellular biliverdin decreases during ferroptosis and that this decrease is specific to ferroptosis amongst different forms of cell death. Furthermore, the feasibility of predicting sensitivity to ferroptosis by measuring intracellular biliverdin was demonstrated using a ferroptosis model induced by the re-expression of the transcription factor BACH1. These findings provide further insight into ferroptosis research and are expected to contribute to the development of cancer therapies that exploit ferroptosis.
Collapse
Affiliation(s)
- Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Guan Chen
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Shunichi Tsuge
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens street, San Francisco, CA 94158, USA
| | - Machi Kiyohara
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Rei Narikawa
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Ohsawa, Hachioji, Tokyo 192-0397, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
11
|
Polito MP, Romaldini A, Tagliazucchi L, Marini G, Radice F, Gozza GA, Bergamini G, Costi MP, Enzo E. Biochemical characterization of the feedforward loop between CDK1 and FOXM1 in epidermal stem cells. Biol Direct 2024; 19:91. [PMID: 39396994 PMCID: PMC11472434 DOI: 10.1186/s13062-024-00540-8] [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: 09/23/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024] Open
Abstract
The complex network governing self-renewal in epidermal stem cells (EPSCs) is only partially defined. FOXM1 is one of the main players in this network, but the upstream signals regulating its activity remain to be elucidated. In this study, we identify cyclin-dependent kinase 1 (CDK1) as the principal kinase controlling FOXM1 activity in human primary keratinocytes. Mass spectrometry identified CDK1 as a key hub in a stem cell-associated protein network, showing its upregulation and interaction with essential self renewal-related markers. CDK1 phosphorylates FOXM1 at specific residues, stabilizing the protein and enhancing its nuclear localization and transcriptional activity, promoting self-renewal. Additionally, FOXM1 binds to the CDK1 promoter, inducing its expression.We identify the CDK1-FOXM1 feedforward loop as a critical axis sustaining EPSCs during in vitro cultivation. Understanding the upstream regulators of FOXM1 activity offers new insights into the biochemical mechanisms underlying self-renewal and differentiation in human primary keratinocytes.
Collapse
Affiliation(s)
- Maria Pia Polito
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Alessio Romaldini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, Modena, 41125, Italy
| | - Grazia Marini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Federica Radice
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Gaia Andrea Gozza
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Giulia Bergamini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, Modena, 41125, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy.
| |
Collapse
|
12
|
Donahue KL, Watkoske HR, Kadiyala P, Du W, Brown K, Scales MK, Elhossiny AM, Espinoza CE, Lasse Opsahl EL, Griffith BD, Wen Y, Sun L, Velez-Delgado A, Renollet NM, Morales J, Nedzesky NM, Baliira RK, Menjivar RE, Medina-Cabrera PI, Rao A, Allen B, Shi J, Frankel TL, Carpenter ES, Bednar F, Zhang Y, Pasca di Magliano M. Oncogenic KRAS-Dependent Stromal Interleukin-33 Directs the Pancreatic Microenvironment to Promote Tumor Growth. Cancer Discov 2024; 14:1964-1989. [PMID: 38958646 PMCID: PMC11450371 DOI: 10.1158/2159-8290.cd-24-0100] [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: 01/30/2024] [Revised: 05/18/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory microenvironment. During carcinogenesis, normal stromal cells are converted to cytokine-high cancer-associated fibroblasts (CAF). The mechanisms underlying this conversion, including the regulation and function of fibroblast-derived cytokines, are poorly understood. Thus, efforts to therapeutically target CAFs have so far failed. Herein, we show that signals from epithelial cells expressing oncogenic KRAS-a hallmark pancreatic cancer mutation-activate fibroblast autocrine signaling, which drives the expression of the cytokine IL33. Stromal IL33 expression remains high and dependent on epithelial KRAS throughout carcinogenesis; in turn, environmental stress induces interleukin-33 (IL33) secretion. Using compartment-specific IL33 knockout mice, we observed that lack of stromal IL33 leads to profound reprogramming of multiple components of the pancreatic tumor microenvironment, including CAFs, myeloid cells, and lymphocytes. Notably, loss of stromal IL33 leads to an increase in CD8+ T-cell infiltration and activation and, ultimately, reduced tumor growth. Significance: This study provides new insights into the mechanisms underlying the programming of CAFs and shows that during this process, expression of the cytokine IL33 is induced. CAF-derived IL33 has pleiotropic effects on the tumor microenvironment, supporting its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Hannah R. Watkoske
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Padma Kadiyala
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan.
| | - Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Kristee Brown
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Michael K. Scales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Ahmed M. Elhossiny
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
| | | | | | | | - Yukang Wen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Lei Sun
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nur M. Renollet
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Jacqueline Morales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nicholas M. Nedzesky
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | | | - Rosa E. Menjivar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.
| | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Cancer Data Science Resource, University of Michigan, Ann Arbor, Michigan.
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan.
| | - Benjamin Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Jiaqi Shi
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, Michigan.
| | - Timothy L. Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Eileen S. Carpenter
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Filip Bednar
- Cancer Biology Program, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
13
|
Dupéré-Richer D, Riva A, Barwick BG, Maji S, Casellas Román H, Li J, De U, Sobh A, Quickstad G, Piper C, Kulis M, Ezponda T, Martín-Subero JI, Tonon G, Zhang W, Mitsiades CS, Boise LH, Bennett RL, Licht JD. KDM6A regulates immune response genes in multiple myeloma. Blood 2024; 144:1508-1520. [PMID: 39046770 PMCID: PMC11952010 DOI: 10.1182/blood.2024024518] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
ABSTRACT The histone H3 at lysine 27 (H3K27) demethylase lysine demethylase 6A (KDM6A) is a tumor suppressor in multiple cancers, including multiple myeloma (MM). We created isogenic MM cells disrupted for KDM6A and tagged the endogenous protein to facilitate genome-wide studies. KDM6A binds genes associated with immune recognition and cytokine signaling. Most importantly, KDM6A binds and activates NLRC5 and CIITA, which encode regulators of major histocompatibility complex genes. Patient data indicate that NLRC5 and CIITA are downregulated in MM with low KDM6A expression. Chromatin analysis shows that KDM6A binds poised and active enhancers and KDM6A loss led to decreased H3K27ac at enhancers, increased H3K27me3 levels in body of genes bound by KDM6A, and decreased gene expression. Reestablishing histone acetylation with an HDAC3 inhibitor leads to upregulation of major histocompatibility complex expression, offering a strategy to restore immunogenicity of KDM6A-deficient tumors. Loss of Kdm6a in Kirsten rat sarcoma virus (K-RAS)-transformed murine fibroblasts led to increased growth in vivo associated with decreased T-cell infiltration.
Collapse
Affiliation(s)
- Daphné Dupéré-Richer
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Alberto Riva
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Winship Cancer Institute, Atlanta, GA
| | - Sayantan Maji
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Heidi Casellas Román
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL
| | - Jianping Li
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Umasankar De
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Amin Sobh
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Gabrielle Quickstad
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Crissandra Piper
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Marta Kulis
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | - Teresa Ezponda
- Hemato-Oncology Department, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Barcelona, Spain
| | - José Ignacio Martín-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain
| | - Giovanni Tonon
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | | | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Winship Cancer Institute, Atlanta, GA
| | - Richard L. Bennett
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| | - Jonathan D. Licht
- Division of Hematology/Oncology, University of Florida Health Cancer Center, University of Florida, Gainesville, FL
| |
Collapse
|
14
|
Velez‐delValle C, Hernandez‐Mosqueira CP, Castro‐Rodriguez LI, Vazquez‐Sandoval A, Marsch‐Moreno M, Kuri‐Harcuch W. Gene expression and characterization of clonally derived murine embryonic brown and brite adipocytes. FEBS Open Bio 2024; 14:1503-1525. [PMID: 38972757 PMCID: PMC11492321 DOI: 10.1002/2211-5463.13861] [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: 01/12/2024] [Revised: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
White adipocytes store energy, while brown and brite adipocytes release heat via nonshivering thermogenesis. In this study, we characterized two murine embryonic clonal preadipocyte lines, EB5 and EB7, each displaying unique gene marker expression profiles. EB5 cells differentiate into brown adipocytes, whereas EB7 cells into brite (also known as beige) adipocytes. To draw a comprehensive comparison, we contrasted the gene expression patterns, adipogenic capacity, as well as carbohydrate and lipid metabolism of these cells to that of F442A, a well-known white preadipocyte and adipocyte model. We found that commitment to differentiation in both EB5 and EB7 cells can be induced by 3-Isobutyl-1-methylxanthine/dexamethasone (Mix/Dex) and staurosporine/dexamethasone (St/Dex) treatments. Additionally, the administration of rosiglitazone significantly enhances the brown and brite adipocyte phenotypes. Our data also reveal the involvement of a series of genes in the transcriptional cascade guiding adipogenesis, pinpointing GSK3β as a critical regulator for both EB5 and EB7 adipogenesis. In a developmental context, we observe that, akin to brown fat progenitors, brite fat progenitors make their appearance in murine development by 11-12 days of gestation or potentially earlier. This result contributes to our understanding of adipocyte lineage specification during embryonic development. In conclusion, EB5 and EB7 cell lines are valuable for research into adipocyte biology, providing insights into the differentiation and development of brown and beige adipocytes. Furthermore, they could be useful for the characterization of drugs targeting energy balance for the treatment of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Cristina Velez‐delValle
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | | | | | | | - Meytha Marsch‐Moreno
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | - Walid Kuri‐Harcuch
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| |
Collapse
|
15
|
Unsal V, Cicek M, Aktepe N, Oner E. Morin attenuates arsenic-induced toxicity in 3T3 embryonic fibroblast cells by suppressing oxidative stress, inflammation, and apoptosis: In vitro and silico evaluations. Toxicol Res (Camb) 2024; 13:tfae113. [PMID: 39036522 PMCID: PMC11260228 DOI: 10.1093/toxres/tfae113] [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: 02/08/2024] [Revised: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
This study aims to investigate the curative effects of Morin, a flavonoid, against arsenic toxicity in 3T3 embryonic fibroblast cells and its effect on the molecular mechanisms of cells. The cytotoxicity and viability of the cells were measured by MTT and LDH tests. Arsenic (0.74 μM) was used to trigger toxicity and Morin (50 μM) was used for treatment. The levels of oxidative stress biomarkers and the activities of antioxidant enzymes were measured by spectrophotometric method, and inflammatory markers were measured by ELISA method. While mRNA expression levels of Bax, Bcl-2 levels, and Caspase-3 activity were measured by qRT-PCR technique, TUNEL staining was performed to detect DNA breaks and DAPI staining to visualize nuclear changes. Protein structures were retrieved from the protein data bank. OpenBabel and Autodock programs were used for the molecular docking study. Morin rescued the 3T3 embryonic fibroblast cells exposed to arsenic. However, Arsenic decreased the activities of antioxidant enzymes in cells and significantly increased oxidative stress, inflammation, and apoptosis. Morin treatment reduced oxidative damage and TNF-α and IL-1β levels. Arsenic-induced Caspase-3 mRNA expression level and Bax protein mRNA expression level were significantly increased, while Bcl-2 mRNA expression level was significantly decreased. While Caspase-3 mRNA expression level and Bax protein mRNA expression level decreased with morin treatment, Bcl-2 mRNA expression level increased significantly. Molecular docking study results showed good binding affinity of morin in SOD, GSH-Px, Bax, Bcl-2, Caspase-3, TNF-α, and IL-1β structures. Morin showed antioxidant, anti-inflammatory, and anti-apoptotic effects against Arsenic-induced cellular toxicity.
Collapse
Affiliation(s)
- Velid Unsal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, Mardin, 47200, Türkiye
| | - Mustafa Cicek
- Department of Medical Biology, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, 46050, Türkiye
| | - Necmettin Aktepe
- Department of Nursing, Faculty of Health Sciences Mardin Artuklu University, Mardin, 47200, Türkiye
| | - Erkan Oner
- Department of Biochemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman, 02000, Türkiye
| |
Collapse
|
16
|
Loaiza-Moss J, Braun U, Leitges M. Transcriptome Profiling of Mouse Embryonic Fibroblast Spontaneous Immortalization: A Comparative Analysis. Int J Mol Sci 2024; 25:8116. [PMID: 39125691 PMCID: PMC11311763 DOI: 10.3390/ijms25158116] [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: 06/26/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Cell immortalization, a hallmark of cancer development, is a process that cells can undergo on their path to carcinogenesis. Spontaneously immortalized mouse embryonic fibroblasts (MEFs) have been used for decades; however, changes in the global transcriptome during this process have been poorly described. In our research, we characterized the poly-A RNA transcriptome changes after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were screened using DESeq2 and characterized by gene ontology enrichment analysis and protein-protein interaction (PPI) network analysis to identify the potential hub genes. In our study, we identified changes in the expression of genes involved in proliferation regulation, cell adhesion, immune response and transcriptional regulation in immortalized MEFs. In addition, we performed a comparative analysis with previously reported MEF immortalization data, where we propose a predicted gene regulatory network model in immortalized MEFs based on the altered expression of Mapk11, Cdh1, Chl1, Zic1, Hoxd10 and the novel hub genes Il6 and Itgb2.
Collapse
Affiliation(s)
| | | | - Michael Leitges
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. Johns, NL A1B 3V6, Canada; (J.L.-M.); (U.B.)
| |
Collapse
|
17
|
Nishizawa H, Matsumoto M, Yamanaka M, Irikura R, Nakajima K, Tada K, Nakayama Y, Konishi M, Itoh N, Funayama R, Nakayama K, Igarashi K. BACH1 inhibits senescence, obesity, and short lifespan by ferroptotic FGF21 secretion. Cell Rep 2024; 43:114403. [PMID: 38943639 DOI: 10.1016/j.celrep.2024.114403] [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: 10/17/2023] [Revised: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron-dependent lipid peroxidation. A model cell system is constructed to induce ferroptosis by re-expressing the transcription factor BACH1, a potent ferroptosis inducer, in immortalized mouse embryonic fibroblasts (iMEFs). The transfer of the culture supernatant from ferroptotic iMEFs activates the proliferation of hepatoma cells and other fibroblasts and suppresses cellular senescence-like features. The BACH1-dependent secretion of the longevity factor FGF21 is increased in ferroptotic iMEFs. The anti-senescent effects of the culture supernatant from these iMEFs are abrogated by Fgf21 knockout. BACH1 activates the transcription of Fgf21 by promoting ferroptotic stress and increases FGF21 protein expression by suppressing its autophagic degradation through transcriptional Sqstm1 and Lamp2 repression. The BACH1-induced ferroptotic FGF21 secretion suppresses obesity in high-fat diet-fed mice and the short lifespan of progeria mice. The inhibition of these aging-related phenotypes can be physiologically significant regarding ferroptosis.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Gladstone Institute of Neurological Disease, Gladstone Institute, San Francisco, CA 94158, USA
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keisuke Tada
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Yoshiaki Nakayama
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Morichika Konishi
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Ryo Funayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keiko Nakayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
18
|
Konishi D, Hirata E, Takano Y, Maeda Y, Ushijima N, Yudasaka M, Yokoyama A. Near-infrared light-boosted antimicrobial activity of minocycline/hyaluronan/carbon nanohorn composite toward peri-implantitis treatments. NANOSCALE 2024; 16:13425-13434. [PMID: 38913014 DOI: 10.1039/d4nr01036a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Dental implant therapy is a reliable treatment for replacing missing teeth. However, as dental implants become more widely used, peri-implantitis increasingly has become a severe complication, making successful treatment more difficult. As a result, the development of effective drug delivery systems (DDSs) and treatments for peri-implantitis are urgently needed. Carbon nanohorns (CNHs) are carbon nanomaterials that have shown promise for use in DDSs and have photothermal effects. The present study exploited the unique properties of CNHs to develop a phototherapy employing a near-infrared (NIR) photoresponsive composite of minocycline, hyaluronan, and CNH (MC/HA/CNH) for peri-implantitis treatments. MC/HA/CNH demonstrated antibacterial effects that were potentiated by NIR-light irradiation, a property that was mediated by photothermal-mediated drug release from HA/CNH. These antibacterial effects persisted even following 48 h of dialysis, a promising indication for the clinical use of this material. We propose that the treatment of peri-implantitis using NIR and MC/HA/CNH, in combination with surgical procedures, might be employed to target relatively deep affected areas in a timely and efficacious manner. We envision that this innovative approach will pave the way for future developments in implant therapy.
Collapse
Affiliation(s)
- Daisuke Konishi
- Department of Oral Functional Prosthodontics, Faculty of Dental Medicine, Graduate school of Dental Medicine, Hokkaido University, Kita-13, Nishi-7, Sapporo 060-8586, Japan.
| | - Eri Hirata
- Department of Oral Functional Prosthodontics, Faculty of Dental Medicine, Graduate school of Dental Medicine, Hokkaido University, Kita-13, Nishi-7, Sapporo 060-8586, Japan.
| | - Yuta Takano
- Research Institute for Electronic Science, Hokkaido University, Kita-20, Nishi-10, Sapporo 001-0020, Japan.
- Graduate School of Environmental Science, Hokkaido University, Kita-10, Nishi-5, Sapporo 060-0810, Japan
| | - Yukari Maeda
- Department of Oral Functional Prosthodontics, Faculty of Dental Medicine, Graduate school of Dental Medicine, Hokkaido University, Kita-13, Nishi-7, Sapporo 060-8586, Japan.
| | - Natsumi Ushijima
- Support Section for Education and Research, Faculty of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan
| | - Masako Yudasaka
- Nanomaterials Research Institute (NMRI), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Meijo University, Graduate School of Science and Technology, 1-501, Shiogamaguchi, Tenpaku, Nagoya 468-8502, Japan
| | - Atsuro Yokoyama
- Department of Oral Functional Prosthodontics, Faculty of Dental Medicine, Graduate school of Dental Medicine, Hokkaido University, Kita-13, Nishi-7, Sapporo 060-8586, Japan.
| |
Collapse
|
19
|
Polito MP, Marini G, Fabrizi A, Sercia L, Enzo E, De Luca M. Biochemical role of FOXM1-dependent histone linker H1B in human epidermal stem cells. Cell Death Dis 2024; 15:508. [PMID: 39019868 PMCID: PMC11255229 DOI: 10.1038/s41419-024-06905-1] [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: 05/31/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Epidermal stem cells orchestrate epidermal renewal and timely wound repair through a tight regulation of self-renewal, proliferation, and differentiation. In culture, human epidermal stem cells generate a clonal type referred to as holoclone, which give rise to transient amplifying progenitors (meroclone and paraclone-forming cells) eventually generating terminally differentiated cells. Leveraging single-cell transcriptomic data, we explored the FOXM1-dependent biochemical signals controlling self-renewal and differentiation in epidermal stem cells aimed at improving regenerative medicine applications. We report that the expression of H1 linker histone subtypes decrease during serial cultivation. At clonal level we observed that H1B is the most expressed isoform, particularly in epidermal stem cells, as compared to transient amplifying progenitors. Indeed, its expression decreases in primary epithelial culture where stem cells are exhausted due to FOXM1 downregulation. Conversely, H1B expression increases when the stem cells compartment is sustained by enforced FOXM1 expression, both in primary epithelial cultures derived from healthy donors and JEB patient. Moreover, we demonstrated that FOXM1 binds the promotorial region of H1B, hence regulates its expression. We also show that H1B is bound to the promotorial region of differentiation-related genes and negatively regulates their expression in epidermal stem cells. We propose a novel mechanism wherein the H1B acts downstream of FOXM1, contributing to the fine interplay between self-renewal and differentiation in human epidermal stem cells. These findings further define the networks that sustain self-renewal along the previously identified YAP-FOXM1 axis.
Collapse
Affiliation(s)
- Maria Pia Polito
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Grazia Marini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Fabrizi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Sercia
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy.
| | - Michele De Luca
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
20
|
Srinivasan S, Henry Ho HY. An efficient method for immortalizing mouse embryonic fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601088. [PMID: 38979230 PMCID: PMC11230376 DOI: 10.1101/2024.06.27.601088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mouse embryonic fibroblasts (MEFs) derived from genetically modified mice are a valuable resource for studying gene function and regulation. The MEF system can also be combined with rescue studies to characterize the function of mutant genes/proteins, such as disease-causing variants. However, primary MEFs undergo senescence soon after isolation and passaging, making long-term genetic manipulations difficult. Previously described methods for MEF immortalization are often inefficient or alter the physiological properties of the cells. Here, we describe an optimized protocol for immortalizing MEFs via CRISPR-mediated deletion of the Tp53 gene. This method is highly efficient and consistently generates immortalized MEFs, or iMEFs, within 14 days. Importantly, iMEFs closely resemble the parent cell populations, and individual iMEFs can be cloned and expanded for subsequent genetic manipulation and characterization. We envision that this protocol can be adopted to immortalize other mouse primary cell types.
Collapse
Affiliation(s)
- Srisathya Srinivasan
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, USA
| |
Collapse
|
21
|
Tsushima H, Tada H, Asai A, Hirose M, Hosoyama T, Watanabe A, Murakami T, Sugimoto M. Roles of pigment epithelium-derived factor in exercise-induced suppression of senescence and its impact on lung pathology in mice. Aging (Albany NY) 2024; 16:10670-10693. [PMID: 38954512 PMCID: PMC11272117 DOI: 10.18632/aging.205976] [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: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Senescent cells contribute to tissue aging and underlie the pathology of chronic diseases. The benefits of eliminating senescent cells have been demonstrated in several disease models, and the efficacy of senolytic drugs is currently being tested in humans. Exercise training has been shown to reduce cellular senescence in several tissues; however, the mechanisms responsible remain unclear. We found that myocyte-derived factors significantly extended the replicative lifespan of fibroblasts, suggesting that myokines mediate the anti-senescence effects of exercise. A number of proteins within myocyte-derived factors were identified by mass spectrometry. Among these, pigment epithelium-derived factor (PEDF) exerted inhibitory effects on cellular senescence. Eight weeks of voluntary running increased Pedf levels in skeletal muscles and suppressed senescence markers in the lungs. The administration of PEDF reduced senescence markers in multiple tissues and attenuated the decline in respiratory function in the pulmonary emphysema mouse model. We also showed that blood levels of PEDF inversely correlated with the severity of COPD in patients. Collectively, these results strongly suggest that PEDF contributes to the beneficial effects of exercise, potentially suppressing cellular senescence and its associated pathologies.
Collapse
Affiliation(s)
- Hiromichi Tsushima
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Hirobumi Tada
- Department of Nutrition, Shigakkan University, Aichi 474-8651, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Azusa Asai
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Mikako Hirose
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Tohru Hosoyama
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Atsushi Watanabe
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| | - Taro Murakami
- Department of Nutrition, Shigakkan University, Aichi 474-8651, Japan
| | - Masataka Sugimoto
- Laboratory of Molecular and Cellular Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan
| |
Collapse
|
22
|
Diaba F, Sandor AG, Morán MDC. Microwave-Assisted Atom Transfer Radical Cyclization in the Synthesis of 3,3-Dichloro-γ- and δ-Lactams from N-Alkenyl-Tethered Trichloroacetamides Catalyzed by RuCl 2(PPh 3) 3 and Their Cytotoxic Evaluation. Molecules 2024; 29:2035. [PMID: 38731527 PMCID: PMC11085086 DOI: 10.3390/molecules29092035] [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: 04/02/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
An expeditious synthesis of γ- and δ-lactams from tethered alkenyl trichloroacetamides in the presence of 5% of RuCl2(PPh3)3 is reported. In this investigation we have demonstrated that microwave activation significantly enhances reaction rates, leading to the formation of the corresponding lactams in yields ranging from good to excellent. Thus, we have been able to prepare a wide range of lactams, including indole and morphan bicyclic scaffolds, where the corresponding reactions were completely diastereoselective. This process was successfully extended to α,α-dichloroamides without affecting either their yield or their diastereoselectivity. Some of the lactams prepared in this work were evaluated for their hemolytic and cytotoxic responses. All compounds were found to be non-hemolytic at the tested concentration, indicating their safety profile in terms of blood cell integrity. Meanwhile, they exhibited interesting cytotoxicity responses that depend on both their lactam structure and cell line. Among the molecules tested, γ-lactam 2a exhibited the lowest IC50 values (100-250 µg/mL) as a function of its cell line, with promising selectivity against squamous carcinoma cells (A431) in comparison with fibroblasts (3T3 cell line).
Collapse
Affiliation(s)
- Faïza Diaba
- Laboratori de Química Orgànica, Facultat de Farmàcia i Ciències de l’Alimentació, IBUB, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Alexandra G. Sandor
- Laboratori de Química Orgànica, Facultat de Farmàcia i Ciències de l’Alimentació, IBUB, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - María del Carmen Morán
- Departament de Bioquímica i Fisiologia-Secció de Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Avda. Joan XXIII 27-31, 08028 Barcelona, Spain;
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
| |
Collapse
|
23
|
Rimal R, Muduli S, Desai P, Marquez AB, Möller M, Platzman I, Spatz J, Singh S. Vascularized 3D Human Skin Models in the Forefront of Dermatological Research. Adv Healthc Mater 2024; 13:e2303351. [PMID: 38277705 PMCID: PMC11468127 DOI: 10.1002/adhm.202303351] [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: 10/02/2023] [Revised: 12/04/2023] [Indexed: 01/28/2024]
Abstract
In vitro engineered skin models are emerging as an alternative platform to reduce and replace animal testing in dermatological research. Despite the progress made in recent years, considerable challenges still exist for the inclusion of diverse cell types within skin models. Blood vessels, in particular, are essential in maintaining tissue homeostasis and are one of many primary contributors to skin disease inception and progression. Substantial efforts in the past have allowed the successful fabrication of vascularized skin models that are currently utilized for disease modeling and drugs/cosmetics testing. This review first discusses the need for vascularization within tissue-engineered skin models, highlighting their role in skin grafting and disease pathophysiology. Second, the review spotlights the milestones and recent progress in the fabrication and utilization of vascularized skin models. Additionally, advances including the use of bioreactors, organ-on-a-chip devices, and organoid systems are briefly explored. Finally, the challenges and future outlook for vascularized skin models are addressed.
Collapse
Affiliation(s)
- Rahul Rimal
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Saradaprasan Muduli
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| | - Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Andrea Bonnin Marquez
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Ilia Platzman
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Joachim Spatz
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
- Max Planck School Matter to LifeJahnstrasse 2969120HeidelbergGermany
| | - Smriti Singh
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| |
Collapse
|
24
|
Lepekhina TB, Nikolaev VV, Darvin ME, Zuhayri H, Snegerev MS, Lozhkomoev AS, Senkina EI, Kokhanenko AP, Lozovoy KA, Kistenev YV. Two-Photon-Excited FLIM of NAD(P)H and FAD-Metabolic Activity of Fibroblasts for the Diagnostics of Osteoimplant Survival. Int J Mol Sci 2024; 25:2257. [PMID: 38396933 PMCID: PMC10889693 DOI: 10.3390/ijms25042257] [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: 12/21/2023] [Revised: 02/04/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Bioinert materials such as the zirconium dioxide and aluminum oxide are widely used in surgery and dentistry due to the absence of cytotoxicity of the materials in relation to the surrounding cells of the body. However, little attention has been paid to the study of metabolic processes occurring at the implant-cell interface. The metabolic activity of mouse 3T3 fibroblasts incubated on yttrium-stabilized zirconium ceramics cured with aluminum oxide (ATZ) and stabilized zirconium ceramics (Y-TZP) was analyzed based on the ratio of the free/bound forms of cofactors NAD(P)H and FAD obtained using two-photon microscopy. The results show that fibroblasts incubated on ceramics demonstrate a shift towards the free form of NAD(P)H, which is observed during the glycolysis process, which, according to our assumptions, is related to the porosity of the surface of ceramic structures. Consequently, despite the high viability and good proliferation of fibroblasts assessed using an MTT test and a scanning electron microscope, the cells are in a state of hypoxia during incubation on ceramic structures. The FLIM results obtained in this work can be used as additional information for scientists who are interested in manufacturing osteoimplants.
Collapse
Affiliation(s)
- Tatiana B. Lepekhina
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
| | - Viktor V. Nikolaev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
| | | | - Hala Zuhayri
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
| | - Mikhail S. Snegerev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
| | - Aleksandr S. Lozhkomoev
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia;
| | - Elena I. Senkina
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
- Institute of Strength Physics and Materials Science of the Siberian Branch of the Russian Academy of Sciences (ISPMS SB RAS), 634021 Tomsk, Russia;
| | - Andrey P. Kokhanenko
- Department of Quantum Electronics and Photonics, Faculty of Radiophysics, National Research Tomsk State University, Lenin Av. 36, 634050 Tomsk, Russia;
| | - Kirill A. Lozovoy
- Department of Quantum Electronics and Photonics, Faculty of Radiophysics, National Research Tomsk State University, Lenin Av. 36, 634050 Tomsk, Russia;
| | - Yury V. Kistenev
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Lenin Ave. 36, 634050 Tomsk, Russia; (T.B.L.); (V.V.N.); (H.Z.); (M.S.S.); (E.I.S.); (Y.V.K.)
| |
Collapse
|
25
|
Dupéré-Richer D, Riva A, Maji S, Barwick BG, Román HC, Sobh A, Quickstad G, Li J, De U, Piper C, Kulis M, Ezponda T, Martin-Subero JI, Tonon G, Zhang W, Mitsiades CS, Boise LH, Bennett RL, Licht JD. KDM6A Regulates Immune Response Genes in Multiple Myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579179. [PMID: 38405853 PMCID: PMC10888870 DOI: 10.1101/2024.02.12.579179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The histone H3K27 demethylase KDM6A is a tumor suppressor in multiple cancers, including multiple myeloma (MM). We created isogenic MM cells disrupted for KDM6A and tagged the endogenous protein to facilitate genome wide studies. KDM6A binds genes associated with immune recognition and cytokine signaling. Most importantly, KDM6A binds and activates NLRC5 and CIITA encoding regulators of Major Histocompatibility Complex (MHC) genes. Patient data indicate that NLRC5 and CIITA, are downregulated in MM with low KDM6A expression. Chromatin analysis shows that KDM6A binds poised and active enhancers and KDM6A loss led to decreased H3K27ac at enhancers, increased H3K27me3 levels in body of genes bound by KDM6A and decreased gene expression. Reestablishing histone acetylation with an HDAC3 inhibitor leads to upregulation of MHC expression, offering a strategy to restore immunogenicity of KDM6A deficient tumors. Loss of Kdm6a in murine RAS-transformed fibroblasts led to increased growth in vivo associated with decreased T cell infiltration. Statement of significance We show that KDM6A participates in immune recognition of myeloma tumor cells by directly regulating the expression of the master regulators of MHC-I and II, NLRC5 and CIITA. The expression of these regulators can by rescued by the HDAC3 inhibitors in KDM6A-null cell lines.
Collapse
|
26
|
Yao J, Liang X, Xu S, Liu Y, Shui L, Li S, Guo H, Xiao Z, Zhao Y, Zheng M. TRAF2 inhibits senescence in hepatocellular carcinoma cells via regulating the ROMO1/ NAD +/SIRT3/SOD2 axis. Free Radic Biol Med 2024; 211:47-62. [PMID: 38043870 DOI: 10.1016/j.freeradbiomed.2023.11.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
The suppression of tumor proliferation via cellular senescence has emerged as a promising approach for anti-tumor therapy. Tumor necrosis factor receptor-associated factor 2 (TRAF2), an adaptor protein involved in the NF-κB signaling pathway and reactive oxygen species (ROS) production, has been implicated in hepatocellular carcinoma (HCC) proliferation. However, little is currently known about whether TRAF2 promotes HCC development by inhibiting cellular senescence. Replicative senescence model and IR-induced mouse model demonstrated that TRAF2 expression was decrease in senescence cells or liver tissues. Depletion of TRAF2 could inhibit proliferation and arrest the cell cycle via activating p53/p21WAF1 and p16INK4a/pRb signaling pathways in HCC cells and eventually lead to cellular senescence. Mechanistically, TRAF2 deficiency increased the expression of mitochondrial protein reactive oxygen species modulator 1 (ROMO1) and subsequently activated the NAD+/SIRT3/SOD2 pathway to promote the production of ROS and cause mitochondrial dysfunction, which eventually contributed to DNA damage response (DDR). Our findings demonstrate that TRAF2 deficiency inhibits the proliferation of HCC by promoting senescence. Therefore, targeting TRAF2 through various approaches holds therapeutic potential for treating HCC.
Collapse
Affiliation(s)
- Jiping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China; Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Xue Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China; Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siduo Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Liyan Shui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Shuangshuang Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Huiting Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Zhengyun Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China
| | - Yongchao Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China; Cancer Center, Zhejiang University, Hangzhou, China; Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, China.
| |
Collapse
|
27
|
Harada M, Su-Harada K, Kimura T, Ono K, Ashida N. Sustained activation of NF-κB through constitutively active IKKβ leads to senescence bypass in murine dermal fibroblasts. Cell Cycle 2024; 23:308-327. [PMID: 38461418 PMCID: PMC11057680 DOI: 10.1080/15384101.2024.2325802] [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: 09/19/2023] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Although the transcription factor nuclear factor κB (NF-κB) plays a central role in the regulation of senescence-associated secretory phenotype (SASP) acquisition, our understanding of the involvement of NF-κB in the induction of cellular senescence is limited. Here, we show that activation of the canonical NF-κB pathway suppresses senescence in murine dermal fibroblasts. IκB kinase β (IKKβ)-depleted dermal fibroblasts showed ineffective NF-κB activation and underwent senescence more rapidly than control cells when cultured under 20% oxygen conditions, as indicated by senescence-associated β-galactosidase (SA-β-gal) staining and p16INK4a mRNA levels. Conversely, the expression of constitutively active IKKβ (IKKβ-CA) was sufficient to drive senescence bypass. Notably, the expression of a degradation-resistant form of inhibitor of κB (IκB), which inhibits NF-κB nuclear translocation, abolished senescence bypass, suggesting that the inhibitory effect of IKKβ-CA on senescence is largely mediated by NF-κB. We also found that IKKβ-CA expression suppressed the derepression of INK4/Arf genes and counteracted the senescence-associated loss of Ezh2, a catalytic subunit of the Polycomb repressive complex 2 (PRC2). Moreover, pharmacological inhibition of Ezh2 abolished IKKβ-CA-induced senescence bypass. We propose that NF-κB plays a suppressive role in the induction of stress-induced senescence through sustaining Ezh2 expression.
Collapse
Affiliation(s)
- Masayuki Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kanae Su-Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noboru Ashida
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
28
|
Gallemí-Pérez A, Tarantola M. Electric Cell-Substrate Impedance Sensing as a Tool to Characterize Wound Healing Dynamics. Methods Mol Biol 2024; 2828:119-145. [PMID: 39147975 DOI: 10.1007/978-1-0716-4023-4_11] [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] [Indexed: 08/17/2024]
Abstract
The electric cell-substrate impedance sensing (ECIS) is a well-established technique that allows for the real-time monitoring of cell cultures growing on gold-electrodes embedded in culture dishes. Its foundation lays on the insulating effect that cells present against the free-flow of electrons, as these passive electrical properties generate a characteristic complex impedance spectrum when a small-amplitude, non-invasive alternating current (AC) is provided through the electrodes, the living cells, and the culture media in the culture ware. In addition, it possesses the ability to create a wound that is highly confined to the electrode area by simply increasing the amplitude of the AC current in dependence of the pre-resistor strength for a defined pulse duration and at a specific frequency. Therefore, it represents a controlled and reproducible tool to carry out in vitro wound healing experiments. Accordingly, in this methods protocol, the use of the ECIS will be described in the context of the wound healing research: cardiac 3T3 fibroblasts will be wounded and their recovery dynamics analyzed based on the typical methodologies applied to the processing of ECIS data. In addition, cellular micromotions will be evaluated. Finally, fluorescence immunostaining of ECIS samples will be described in order to showcase the potential of the ECIS in combination with other well-established techniques to add further knowledge depth to the understanding of the complex wound healing dynamics.
Collapse
Affiliation(s)
- Aina Gallemí-Pérez
- Max Planck Institute for Dynamics and Self-Organisation, Göttingen, Germany.
| | - Marco Tarantola
- Max Planck Institute for Dynamics and Self-Organisation, Göttingen, Germany
| |
Collapse
|
29
|
Kizilirmak C, Monteleone E, García-Manteiga JM, Brambilla F, Agresti A, Bianchi ME, Zambrano S. Small transcriptional differences among cell clones lead to distinct NF-κB dynamics. iScience 2023; 26:108573. [PMID: 38144455 PMCID: PMC10746373 DOI: 10.1016/j.isci.2023.108573] [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: 06/14/2023] [Revised: 10/06/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
Transcription factor dynamics is fundamental to determine the activation of accurate transcriptional programs and yet is heterogeneous at a single-cell level, even within homogeneous populations. We asked how such heterogeneity emerges for the nuclear factor κB (NF-κB). We found that clonal populations of immortalized fibroblasts derived from a single mouse embryo display robustly distinct NF-κB dynamics upon tumor necrosis factor ɑ (TNF-ɑ) stimulation including persistent, oscillatory, and weak activation, giving rise to differences in the transcription of its targets. By combining transcriptomics and simulations we show how less than two-fold differences in the expression levels of genes coding for key proteins of the signaling cascade and feedback system are predictive of the differences of the NF-κB dynamic response of the clones to TNF-ɑ and IL-1β. We propose that small transcriptional differences in the regulatory circuit of a transcription factor can lead to distinct signaling dynamics in cells within homogeneous cell populations and among different cell types.
Collapse
Affiliation(s)
- Cise Kizilirmak
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Emanuele Monteleone
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Francesca Brambilla
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandra Agresti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco E. Bianchi
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Samuel Zambrano
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
30
|
Yang L, Jing Y, Xia X, Yin X. ARV-825 Showed Antitumor Activity against BRD4-NUT Fusion Protein by Targeting the BRD4. JOURNAL OF ONCOLOGY 2023; 2023:9904143. [PMID: 38130463 PMCID: PMC10735731 DOI: 10.1155/2023/9904143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 10/07/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Objective The bromodomain-containing 4 (BRD4) is a member of the bromodomain and extra terminal domain (BET) family, which is an important epigenetic reader. It is currently a promising oncology target. In some tumors, BET bromodomain inhibitors have demonstrated promising results. Proteolysis-targeting methods (PROTAC), which rapidly and effectively degrade BRD4, have displayed considerable potential in the treatment of tumors in recent years. The purpose of this study is to examine the potential impact of BRD4 PROTAC compounds ARV-825 on oncogene BRD4-NUT fused protein in NUT carcinoma. Methods The effectiveness of ARV-825 was evaluated at the cellular level using the cell counting kit 8 test, wound healing, cell transfection, western blotting analysis, and RNA sequencing. The effectiveness of ARV-825 was also examined in vivo using a xenograft model. Results The BRD4-NUT fusion gene was overexpressed in 3T3 cells, and the pathogenic fusion gene was simulated. The results showed that the overexpression of BRD4-NUT could promote the proliferation and migration of 3T3 cells, but the expression of BRD4 protein was degraded after the addition of the novel cereblon-based PROTAC compound ARV-825 against BRD4, resulting in inhibition of BRD4-NUT 3T3 cell proliferation and migration. Further RNA-seq analysis showed that overexpression of BRD4-NUT was accompanied by increased expression of gene (e.g., Myc, E2F, TRAFs, Wnt, Gadd45g, and Sox6) with significantly enriched pathway (e.g., small cell lung cancer, NF-kappa B signaling pathway, and breast cancer), promoted cell cycle from G 1 phase to S phase, and increased cell proliferation and migration, activated the antiapoptosisi signal, led to abnormal cell growth, and ultimately led to tumorigenesis. The addition of ARV-825 effectively rescued this process and effectively inhibited cell vitality, proliferation, and migration. In vivo studies demonstrated that treatment with ARV-825 greatly suppressed tumor growth without causing harmful side effects and downregulated the BRD4-NUT expression level. Conclusion Through the induction of BRD4 protein degradation, ARV-825 can successfully limit BRD4-NUT 3T3 cell proliferation in vitro and in vivo. These findings suggested that the BRD4 inhibitor ARV-825 would be an effective therapeutic strategy for treating NUT carcinoma that with the genetic feature of BRD4-NUT fusion event.
Collapse
Affiliation(s)
- Liu Yang
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yue Jing
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Xia Xia
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Roc Rock Biotechnology (Shenzhen), Shenzhen 518118, China
| |
Collapse
|
31
|
Braillard S, Keenan M, Breese KJ, Heppell J, Abbott M, Islam R, Shackleford DM, Katneni K, Crighton E, Chen G, Patil R, Lee G, White KL, Carvalho S, Wall RJ, Chemi G, Zuccotto F, González S, Marco M, Deakyne J, Standing D, Brunori G, Lyon JJ, Castañeda Casado P, Camino I, Martinez MSM, Zulfiqar B, Avery VM, Feijens PB, Van Pelt N, Matheeussen A, Hendrickx S, Maes L, Caljon G, Yardley V, Wyllie S, Charman SA, Chatelain E. DNDI-6174 is a preclinical candidate for visceral leishmaniasis that targets the cytochrome bc 1. Sci Transl Med 2023; 15:eadh9902. [PMID: 38091406 PMCID: PMC7615677 DOI: 10.1126/scitranslmed.adh9902] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/12/2023] [Indexed: 12/18/2023]
Abstract
New drugs for visceral leishmaniasis that are safe, low cost, and adapted to the field are urgently required. Despite concerted efforts over the last several years, the number of new chemical entities that are suitable for clinical development for the treatment of Leishmania remains low. Here, we describe the discovery and preclinical development of DNDI-6174, an inhibitor of Leishmania cytochrome bc1 complex activity that originated from a phenotypically identified pyrrolopyrimidine series. This compound fulfills all target candidate profile criteria required for progression into preclinical development. In addition to good metabolic stability and pharmacokinetic properties, DNDI-6174 demonstrates potent in vitro activity against a variety of Leishmania species and can reduce parasite burden in animal models of infection, with the potential to approach sterile cure. No major flags were identified in preliminary safety studies, including an exploratory 14-day toxicology study in the rat. DNDI-6174 is a cytochrome bc1 complex inhibitor with acceptable development properties to enter preclinical development for visceral leishmaniasis.
Collapse
Affiliation(s)
- Stéphanie Braillard
- Drugs for Neglected Diseases initiative (DNDi), Chemin Camille-Vidart 15, 1202 Geneva, Switzerland
| | | | | | - Jacob Heppell
- Epichem Pty Ltd, Perth, Western Australia, Australia
| | | | - Rafiqul Islam
- Epichem Pty Ltd, Perth, Western Australia, Australia
| | - David M. Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Rahul Patil
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Given Lee
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Karen L. White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Sandra Carvalho
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Richard J. Wall
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Giulia Chemi
- Drug Discovery Unit, Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Fabio Zuccotto
- Drug Discovery Unit, Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Silvia González
- Global Health Medicines R&D, GlaxoSmithKline, Tres Cantos, Madrid 28760, Spain
| | - Maria Marco
- Global Health Medicines R&D, GlaxoSmithKline, Tres Cantos, Madrid 28760, Spain
| | | | | | - Gino Brunori
- Global Investigative Safety, GSK, Ware, United Kingdom
| | | | | | | | | | - Bilal Zulfiqar
- Discovery Biology, Griffith University, Nathan, Queensland, Australia 4111
| | - Vicky M. Avery
- Discovery Biology, Griffith University, Nathan, Queensland, Australia 4111
| | - Pim-Bart Feijens
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Natascha Van Pelt
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Vanessa Yardley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Susan Wyllie
- Wellcome Centre for Anti-infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative (DNDi), Chemin Camille-Vidart 15, 1202 Geneva, Switzerland
| |
Collapse
|
32
|
Lewis AM, Foseh G, Tu W, Peden K, Akue A, KuKuruga M, Rotroff D, Lewis G, Mazo I, Bauer SR. GLI1+ perivascular, renal, progenitor cells: The likely source of spontaneous neoplasia that created the AGMK1-9T7 cell line. PLoS One 2023; 18:e0293406. [PMID: 38060571 PMCID: PMC10703308 DOI: 10.1371/journal.pone.0293406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/11/2023] [Indexed: 12/18/2023] Open
Abstract
The AGMK1-9T7 cell line has been used to study neoplasia in tissue culture. By passage in cell culture, these cells evolved to become tumorigenic and metastatic in immunodeficient mice at passage 40. Of the 20 x 106 kidney cells originally plated, less than 2% formed the colonies that evolved to create this cell line. These cells could be the progeny of some type of kidney progenitor cells. To characterize these cells, we documented their renal lineage by their expression of PAX-2 and MIOX, detected by indirect immunofluorescence. These cells assessed by flow-cytometry expressed high levels of CD44, CD73, CD105, Sca-1, and GLI1 across all passages tested; these markers have been reported to be expressed by renal progenitor cells. The expression of GLI1 was confirmed by immunofluorescence and western blot analysis. Cells from passages 13 to 23 possessed the ability to differentiate into adipocytes, osteoblasts, and chondrocytes; after passage 23, their ability to form these cell types was lost. These data indicate that the cells that formed the AGMK1-9T7 cell line were GLI1+ perivascular, kidney, progenitor cells.
Collapse
Affiliation(s)
- Andrew M. Lewis
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Gideon Foseh
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Wei Tu
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Keith Peden
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Adovi Akue
- Flow Cytometry Unit, OMPT, Center for Biologics Evaluation and Research, OVRR, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Mark KuKuruga
- Flow Cytometry Unit, OMPT, Center for Biologics Evaluation and Research, OVRR, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Daniel Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Gladys Lewis
- TCL and M Associates, Leesburg, Virginia, United States of America
| | - Ilya Mazo
- HIVE Team, Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, FDA, Silver Spring, Maryland, United States of America
| | - Steven R. Bauer
- Division of Cellular and Gene Therapies, Office of Therapeutic Products, Center for Biologics Evaluation and Research, Silver Spring, Maryland, United States of America
| |
Collapse
|
33
|
Hong TJ, Sivakumar C, Luo CW, Ho MS. Investigation of TiO 2 nanoparticle interactions in the fibroblast NIH-3T3 cells via liquid-mode atomic force microscope. Arch Toxicol 2023; 97:2893-2901. [PMID: 37612376 DOI: 10.1007/s00204-023-03585-2] [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: 06/12/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023]
Abstract
Long before we recognized how significant they were, nanoparticles were already all around in the environment. Since then, an extensive number of synthetic nanoparticles have been engineered to improve our quality of life through rigorous scientific research on their uses in practically every industry, including semiconductor devices, food, medicine, and agriculture. The extensive usage of nanoparticles in commodities that come into proximity with human skin and internal organs through medicine has raised significant concerns over the years. TiO2 nanoparticles (NPs) are widely employed in a wide range of industries, such as cosmetics and food packaging. The interaction and internalization of TiO2 NPs in living cells have been studied by the scientific community for many years. In the present study, we investigated the cell viability, nanomechanical characteristics, and fluorescence response of NIH-3T3 cells treated with sterile DMEM TiO2 nanoparticle solution using a liquid-mode atomic force microscope and a fluorescence microscope. Two different sorts of response systems have been observed in the cells depending on the size of the NPs. TiO2 nanoparticles smaller than 100 nm support its initial stages cell viability, and cells internalize and metabolize NPs. In contrast, bigger TiO2 NPs (> 100 nm) are not completely metabolized and cannot impair cell survival. Furthermore, bigger NPs above 100 nm could not be digested by the cells, therefore hindering cell development, whereas below 100 nm TiO2 stimulated uncontrolled cell growth akin to cancerous type cells. The cytoskeleton softens as a result of particle internalization, as seen by the nanomechanical characteristics of the nanoparticle treated cells. According to our investigations, TiO2 smaller than 100 nm facilitates unintended cancer cell proliferation, whereas larger NPs ultimately suppress cell growth. Before being incorporated into commercial products, similar effects or repercussions that could result from employing different NPs should be carefully examined.
Collapse
Affiliation(s)
- Tz-Ju Hong
- Department of Electrophysics, National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan
| | | | - Chih-Wei Luo
- Department of Electrophysics, National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan
- National Synchrotron Radiation Research Center (NSRRC), Hsinchu, 30076, Taiwan
- Institute of Physics and Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University, Hsinchu, 30010, Taiwan
- Taiwan Consortium of Emergent Crystalline Materials (TCECM), Ministry of Science and Technology, Taipei, 10601, Taiwan
| | - Mon-Shu Ho
- Department of Physics, National Chung Hsing University, Taichung City, 40227, Taiwan.
| |
Collapse
|
34
|
Abstract
Mouse embryonic fibroblasts (MEFs) are primary fibroblasts purified from mouse embryos at a defined time post-fertilization. MEFs have versatile applications, including use as feeder cell layers or sources of untransformed primary cells for a variety of biological assays. MEFs are most commonly isolated between embryonic day (E)12.5 and E13.5 but can be isolated from embryos as early as E8.5 and as late as E15.5. The individual embryos are harvested by carefully removing uterine tissue, yolk sac, and placenta. The embryos are euthanized, and non-mesenchymal tissues, such as the fetal liver and heart, are removed before tissue homogenization. The remaining fetal tissue is homogenized by mechanical mincing using a sterile blade, followed by enzymatic digestion and resuspension. During tissue dissociation, the duration of trypsin-EDTA/DNase digestion and enzyme concentration are critical parameters to produce high-quality MEFs with the highest rates of cell viability and proliferation potential. MEFs can be cryopreserved at passage (P) 0 if >80% confluent, passaged for further expansion before freezing down, or directly utilized for downstream applications, i.e., preparation as feeder cell layers. Primary MEFs possess a limited proliferation capacity of ∼20 cell divisions, beyond which the percentage of senescent cells rapidly increases; thus, cultures should only be expanded/passaged to a maximum of P5. Critical for cell viability during cryopreservation and thawing of MEFs is the slow decrease in temperature when freezing, the rapid increase when thawing, the use of a cryoprotective agent, and an optimal cell density. While it is critical to generate high-quality MEFs to standardize and optimize preparation procedures and utilize fresh reagents, some variability in proliferation capacity and cell viability between MEF preparations remains. Thus, MEF preparation, culture, and cryopreservation procedures are continuously being optimized. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Purification, passaging, and expansion of MEFs Supporting Protocol: Cryopreservation and thawing of MEFs.
Collapse
Affiliation(s)
- Rita Ferreira
- ACRF Department of Cancer Biology and Therapeutics and Division of Genome Sciences and Cancer, John Curtin School of Medical Research, Australian National University, Acton, Canberra, Australia
| | - Nadine Hein
- ACRF Department of Cancer Biology and Therapeutics and Division of Genome Sciences and Cancer, John Curtin School of Medical Research, Australian National University, Acton, Canberra, Australia
| |
Collapse
|
35
|
Hay AD, Kessler NJ, Gebert D, Takahashi N, Tavares H, Teixeira FK, Ferguson-Smith AC. Epigenetic inheritance is unfaithful at intermediately methylated CpG sites. Nat Commun 2023; 14:5336. [PMID: 37660134 PMCID: PMC10475082 DOI: 10.1038/s41467-023-40845-2] [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: 03/14/2023] [Accepted: 08/12/2023] [Indexed: 09/04/2023] Open
Abstract
DNA methylation at the CpG dinucleotide is considered a stable epigenetic mark due to its presumed long-term inheritance through clonal expansion. Here, we perform high-throughput bisulfite sequencing on clonally derived somatic cell lines to quantitatively measure methylation inheritance at the nucleotide level. We find that although DNA methylation is generally faithfully maintained at hypo- and hypermethylated sites, this is not the case at intermediately methylated CpGs. Low fidelity intermediate methylation is interspersed throughout the genome and within genes with no or low transcriptional activity, and is not coordinately maintained between neighbouring sites. We determine that the probabilistic changes that occur at intermediately methylated sites are likely due to DNMT1 rather than DNMT3A/3B activity. The observed lack of clonal inheritance at intermediately methylated sites challenges the current epigenetic inheritance model and has direct implications for both the functional relevance and general interpretability of DNA methylation as a stable epigenetic mark.
Collapse
Affiliation(s)
- Amir D Hay
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Noah J Kessler
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Daniel Gebert
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Nozomi Takahashi
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Hugo Tavares
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Felipe K Teixeira
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK.
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK.
| |
Collapse
|
36
|
Kazyken D, Lentz SI, Wadley M, Fingar DC. Alkaline intracellular pH (pHi) increases PI3K activity to promote mTORC1 and mTORC2 signaling and function during growth factor limitation. J Biol Chem 2023; 299:105097. [PMID: 37507012 PMCID: PMC10477693 DOI: 10.1016/j.jbc.2023.105097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The conserved protein kinase mTOR (mechanistic target of rapamycin) responds to diverse environmental cues to control cell metabolism and promote cell growth, proliferation, and survival as part of two multiprotein complexes, mTOR complex 1 (mTORC1) and mTORC2. Our prior work demonstrated that an alkaline intracellular pH (pHi) increases mTORC2 activity and cell survival in complete media in part by activating AMP-activated protein kinase, a kinase best known to sense energetic stress. It is important to note that an alkaline pHi represents an underappreciated hallmark of cancer cells that promotes their oncogenic behaviors. In addition, mechanisms that control mTORC1 and mTORC2 signaling and function remain incompletely defined, particularly in response to stress conditions. Here, we demonstrate that an alkaline pHi increases phosphatidylinositide 3-kinase (PI3K) activity to promote mTORC1 and mTORC2 signaling in the absence of serum growth factors. Alkaline pHi increases mTORC1 activity through PI3K-Akt signaling, which mediates inhibitory phosphorylation of the upstream proteins tuberous sclerosis complex 2 and proline-rich Akt substrate of 40 kDa and dissociates tuberous sclerosis complex from lysosomal membranes, thus enabling Rheb-mediated activation of mTORC1. Thus, alkaline pHi mimics growth factor-PI3K signaling. Functionally, we also demonstrate that an alkaline pHi increases cap-dependent protein synthesis through inhibitory phosphorylation of eIF4E binding protein 1 and suppresses apoptosis in a PI3K- and mTOR-dependent manner. We speculate that an alkaline pHi promotes a low basal level of cell metabolism (e.g., protein synthesis) that enables cancer cells within growing tumors to proliferate and survive despite limiting growth factors and nutrients, in part through elevated PI3K-mTORC1 and/or PI3K-mTORC2 signaling.
Collapse
Affiliation(s)
- Dubek Kazyken
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Stephen I Lentz
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maxwell Wadley
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Diane C Fingar
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
37
|
Irikura R, Nishizawa H, Nakajima K, Yamanaka M, Chen G, Tanaka K, Onodera M, Matsumoto M, Igarashi K. Ferroptosis model system by the re-expression of BACH1. J Biochem 2023; 174:239-252. [PMID: 37094356 DOI: 10.1093/jb/mvad036] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
Ferroptosis is a regulated cell death induced by iron-dependent lipid peroxidation. The heme-responsive transcription factor BTB and CNC homology 1 (BACH1) promotes ferroptosis by repressing the transcription of genes involved in glutathione (GSH) synthesis and intracellular labile iron metabolism, which are key regulatory pathways in ferroptosis. We found that BACH1 re-expression in Bach1-/- immortalized mouse embryonic fibroblasts (iMEFs) can induce ferroptosis upon 2-mercaptoethanol removal, without any ferroptosis inducers. In these iMEFs, GSH synthesis was reduced, and intracellular labile iron levels were increased upon BACH1 re-expression. We used this system to investigate whether the major ferroptosis regulators glutathione peroxidase 4 (Gpx4) and apoptosis-inducing factor mitochondria-associated 2 (Aifm2), the gene for ferroptosis suppressor protein 1, are target genes of BACH1. Neither Gpx4 nor Aifm2 was regulated by BACH1 in the iMEFs. However, we found that BACH1 represses AIFM2 transcription in human pancreatic cancer cells. These results suggest that the ferroptosis regulators targeted by BACH1 may vary across different cell types and animal species. Furthermore, we confirmed that the ferroptosis induced by BACH1 re-expression exhibited a propagating effect. BACH1 re-expression represents a new strategy for inducing ferroptosis after GPX4 or system Xc- suppression and is expected to contribute to future ferroptosis research.
Collapse
Key Words
- BACH1 Abbreviations: AIFM2, apoptosis-inducing factor mitochondria-associated 2; ANOVA, analysis of variance; BACH1, BTB and CNC homology 1; Bach1−/− mice, Bach1 knockout mice; BTB, Broad complex, Tramtrack, Bric-a-brac domain; bZIP, basic leucine zipper; ChIP-seq, chromatin immunoprecipitation sequencing; CNC, Cap‘n’Collar region; DAPI, 4′,6-diamidino-2-phenylindole; DFX, deferasirox; DMSO, dimethyl sulfoxide; EMT, epithelial–mesenchymal transition; Ferr-1, ferrostatin-1; FINs, ferroptosis inducers; FSP1, Ferroptosis suppressor protein 1; Fth1, ferritin heavy chain 1; Ftl, ferritin light chain; GCL, glutamate-cysteine ligase; Gclc, GCL catalytic subunit; Gclm, GCL modifier subunit; GEO, Gene Expression Omnibus; GPX4, glutathione peroxidase 4; GSH, glutathione; HO-1 (Hmox1), heme oxygenase 1; iMEFs, immortalized MEFs; KuO, Kusabira Orange; MAFK, musculoaponeurotic fibrosarcoma oncogene homolog bZIP transcription factor K; mBACH1, Bach1 gene of Mus musculus; 2-ME, 2-mercaptoethanol; MEFs, mouse embryonic fibroblasts; NRF2, nuclear factor-erythroid 2-related factor 2; NSA, necrosulfonamide; PDAC, pancreatic ductal adenocarcinoma; PI, Propidium iodide; Ptgs2, prostaglandin-endoperoxide synthase 2; RSL3, (1S,3R)-RSL3; Slc40a1, solute carrier family 40 member 1; Slc7a11, solute carrier family 7 member 11; TFRC, transferrin receptor 1; Z-VAD.FMK, Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone
- extracellular signal
- ferroptosis
- fibroblasts
- transcription
Collapse
Affiliation(s)
- Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Guan Chen
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Masafumi Onodera
- Gene & Cell Therapy Promotion Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
38
|
Du H, Xiao N, Zhang S, Zhou X, Zhang Y, Lu Z, Fu Y, Huang M, Xu S, Chen Q. Suppression of TREX1 deficiency-induced cellular senescence and interferonopathies by inhibition of DNA damage response. iScience 2023; 26:107090. [PMID: 37416470 PMCID: PMC10320204 DOI: 10.1016/j.isci.2023.107090] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/16/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
TREX1 encodes a major DNA exonuclease and mutations of this gene are associated with type I interferonopathies in human. Mice with Trex1 deletion or mutation have shortened life spans accompanied by a senescence-associated secretory phenotype. However, the contribution of cellular senescence in TREX1 deficiency-induced type I interferonopathies remains unknown. We found that features of cellular senescence present in Trex1-/- mice are induced by multiple factors, particularly DNA damage. The cGAS-STING and DNA damage response pathways are required for maintaining TREX1 deletion-induced cellular senescence. Inhibition of the DNA damage response, such as with Checkpoint kinase 2 (CHK2) inhibitor, partially alleviated progression of type I interferonopathies and lupus-like features in the mice. These data provide insights into the initiation and development of type I interferonopathies and lupus-like diseases, and may help inform the development of targeted therapeutics.
Collapse
Affiliation(s)
- Hekang Du
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Nanyang Xiao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
- Department of Microbiology, University of Chicago, Cummings Life Science Center, 920 East 58th Street, Chicago, IL 60637, USA
| | - Sitong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Xueyuan Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Yangfan Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Zengzeng Lu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Yuqian Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Miaohui Huang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, China
| |
Collapse
|
39
|
Dill MN, Tabatabaei M, Kamat M, Basso KB, Moore E, Simmons CS. Generation and characterization of two immortalized dermal fibroblast cell lines from the spiny mouse (Acomys). PLoS One 2023; 18:e0280169. [PMID: 37418364 PMCID: PMC10328323 DOI: 10.1371/journal.pone.0280169] [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: 12/21/2022] [Accepted: 06/14/2023] [Indexed: 07/09/2023] Open
Abstract
The spiny mouse (Acomys) is gaining popularity as a research organism due to its phenomenal regenerative capabilities. Acomys recovers from injuries to several organs without fibrosis. For example, Acomys heals full thickness skin injuries with rapid re-epithelialization of the wound and regeneration of hair follicles, sebaceous glands, erector pili muscles, adipocytes, and dermis without scarring. Understanding mechanisms of Acomys regeneration may uncover potential therapeutics for wound healing in humans. However, access to Acomys colonies is limited and primary fibroblasts can only be maintained in culture for a limited time. To address these obstacles, we generated immortalized Acomys dermal fibroblast cell lines using two methods: transfection with the SV40 large T antigen and spontaneous immortalization. The two cell lines (AcoSV40 and AcoSI-1) maintained the morphological and functional characteristics of primary Acomys fibroblasts, including maintenance of key fibroblast markers and ECM deposition. The availability of these cells will lower the barrier to working with Acomys as a model research organism, increasing the pace at which new discoveries to promote regeneration in humans can be made.
Collapse
Affiliation(s)
- Michele N. Dill
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Mohammad Tabatabaei
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Manasi Kamat
- Department of Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Kari B. Basso
- Department of Chemistry, University of Florida, Gainesville, Florida, United States of America
| | - Erika Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Chelsey S. Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States of America
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
40
|
Alkhoury C, Henneman NF, Petrenko V, Shibayama Y, Segaloni A, Gadault A, Nemazanyy I, Le Guillou E, Wolide AD, Antoniadou K, Tong X, Tamaru T, Ozawa T, Girard M, Hnia K, Lutter D, Dibner C, Panasyuk G. Class 3 PI3K coactivates the circadian clock to promote rhythmic de novo purine synthesis. Nat Cell Biol 2023; 25:975-988. [PMID: 37414850 PMCID: PMC10344785 DOI: 10.1038/s41556-023-01171-3] [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: 05/30/2022] [Accepted: 05/22/2023] [Indexed: 07/08/2023]
Abstract
Metabolic demands fluctuate rhythmically and rely on coordination between the circadian clock and nutrient-sensing signalling pathways, yet mechanisms of their interaction remain not fully understood. Surprisingly, we find that class 3 phosphatidylinositol-3-kinase (PI3K), known best for its essential role as a lipid kinase in endocytosis and lysosomal degradation by autophagy, has an overlooked nuclear function in gene transcription as a coactivator of the heterodimeric transcription factor and circadian driver Bmal1-Clock. Canonical pro-catabolic functions of class 3 PI3K in trafficking rely on the indispensable complex between the lipid kinase Vps34 and regulatory subunit Vps15. We demonstrate that although both subunits of class 3 PI3K interact with RNA polymerase II and co-localize with active transcription sites, exclusive loss of Vps15 in cells blunts the transcriptional activity of Bmal1-Clock. Thus, we establish non-redundancy between nuclear Vps34 and Vps15, reflected by the persistent nuclear pool of Vps15 in Vps34-depleted cells and the ability of Vps15 to coactivate Bmal1-Clock independently of its complex with Vps34. In physiology we find that Vps15 is required for metabolic rhythmicity in liver and, unexpectedly, it promotes pro-anabolic de novo purine nucleotide synthesis. We show that Vps15 activates the transcription of Ppat, a key enzyme for the production of inosine monophosphate, a central metabolic intermediate for purine synthesis. Finally, we demonstrate that in fasting, which represses clock transcriptional activity, Vps15 levels are decreased on the promoters of Bmal1 targets, Nr1d1 and Ppat. Our findings open avenues for establishing the complexity for nuclear class 3 PI3K signalling for temporal regulation of energy homeostasis.
Collapse
Affiliation(s)
- Chantal Alkhoury
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Nathaniel F Henneman
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Volodymyr Petrenko
- The Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Yui Shibayama
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Arianna Segaloni
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Alexis Gadault
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS, UAR 3633, Paris, France
| | - Edouard Le Guillou
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Amare Desalegn Wolide
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München (TUM), Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Konstantina Antoniadou
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Xin Tong
- Department of Molecular and Integrative Physiology, Caswell Diabetes Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Teruya Tamaru
- Department of Physiology, Toho University School of Medicine, Tokyo, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Muriel Girard
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM U1151/CNRS UMR 8253, Paris, France
- Université Paris Cité, Paris, France
| | - Karim Hnia
- Institute of Cardiovascular and Metabolic Diseases (I2MC), INSERM-UMR 1297, University Paul Sabatier, Toulouse, France
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Charna Dibner
- The Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Ganna Panasyuk
- Institut Necker-Enfants Malades (INEM), Paris, France.
- INSERM U1151/CNRS UMR 8253, Paris, France.
- Université Paris Cité, Paris, France.
| |
Collapse
|
41
|
Dzhambazov B, Batsalova T, Merky P, Lange F, Holmdahl R. NIH/3T3 Fibroblasts Selectively Activate T Cells Specific for Posttranslationally Modified Collagen Type II. Int J Mol Sci 2023; 24:10811. [PMID: 37445989 DOI: 10.3390/ijms241310811] [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: 06/06/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
It has been shown that synovial fibroblasts (SF) play a key role in the initiation of inflammation and joint destruction, leading to arthritis progression. Fibroblasts may express major histocompatibility complex class II region (MHCII) molecules, and thus, they could be able to process and present antigens to immunocompetent cells. Here we examine whether different types of fibroblasts (synovial, dermal, and thymic murine fibroblasts, destructive LS48 fibroblasts, and noninvasive NIH/3T3 fibroblasts) may be involved in the initiation of rheumatoid arthritis (RA) pathogenesis and can process and present type II collagen (COL2)-an autoantigen associated with RA. Using a panel of MHCII/Aq-restricted T-cell hybridoma lines that specifically recognize an immunodominant COL2 epitope (COL2259-273), we found that NIH/3T3 fibroblasts activate several T-cell clones that recognize the posttranslationally glycosylated or hydroxylated COL2259-273 epitope. The HCQ.3 hybridoma, which is specific for the glycosylated immunodominant COL2 epitope 259-273 (Gal264), showed the strongest response. Interestingly, NIH/3T3 cells, but not destructive LS48 fibroblasts, synovial, dermal, or thymic fibroblasts, were able to stimulate the HCQ.3 hybridoma and other COL2-specific T-cell hybridomas. Our experiments revealed that NIH/3T3 fibroblasts are able to activate COL2-specific T-cell hybridomas even in the absence of COL2 or a posttranslationally modified COL2 peptide. The mechanism of this unusual activation is contact-dependent and involves the T-cell receptor (TCR) complex.
Collapse
Affiliation(s)
- Balik Dzhambazov
- Faculty of Biology, Paisii Hilendarski University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Tsvetelina Batsalova
- Faculty of Biology, Paisii Hilendarski University of Plovdiv, 4000 Plovdiv, Bulgaria
| | | | - Franziska Lange
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
| | - Rikard Holmdahl
- Section of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| |
Collapse
|
42
|
Arena GO, Forte S, Abdouh M, Vanier C, Corbeil D, Lorico A. Horizontal Transfer of Malignant Traits and the Involvement of Extracellular Vesicles in Metastasis. Cells 2023; 12:1566. [PMID: 37371036 PMCID: PMC10297028 DOI: 10.3390/cells12121566] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Metastases are responsible for the vast majority of cancer deaths, yet most therapeutic efforts have focused on targeting and interrupting tumor growth rather than impairing the metastatic process. Traditionally, cancer metastasis is attributed to the dissemination of neoplastic cells from the primary tumor to distant organs through blood and lymphatic circulation. A thorough understanding of the metastatic process is essential to develop new therapeutic strategies that improve cancer survival. Since Paget's original description of the "Seed and Soil" hypothesis over a hundred years ago, alternative theories and new players have been proposed. In particular, the role of extracellular vesicles (EVs) released by cancer cells and their uptake by neighboring cells or at distinct anatomical sites has been explored. Here, we will outline and discuss these alternative theories and emphasize the horizontal transfer of EV-associated biomolecules as a possibly major event leading to cell transformation and the induction of metastases. We will also highlight the recently discovered intracellular pathway used by EVs to deliver their cargoes into the nucleus of recipient cells, which is a potential target for novel anti-metastatic strategies.
Collapse
Affiliation(s)
- Goffredo O. Arena
- Department of Surgery, McGill University, Montréal, QC H3A 0G4, Canada;
- Fondazione Istituto G. Giglio, 90015 Cefalù, Italy
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute, McGill University Health Centre, Montréal, QC H3A 0G4, Canada;
| | - Cheryl Vanier
- Touro University Nevada College of Medicine, Henderson, NV 89014, USA;
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Aurelio Lorico
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy;
- Touro University Nevada College of Medicine, Henderson, NV 89014, USA;
| |
Collapse
|
43
|
Li Z, Jiao X, Robertson AG, Di Sante G, Ashton AW, DiRocco A, Wang M, Zhao J, Addya S, Wang C, McCue PA, South AP, Cordon-Cardo C, Liu R, Patel K, Hamid R, Parmar J, DuHadaway JB, Jones SJM, Casimiro MC, Schultz N, Kossenkov A, Phoon LY, Chen H, Lan L, Sun Y, Iczkowski KA, Rui H, Pestell RG. The DACH1 gene is frequently deleted in prostate cancer, restrains prostatic intraepithelial neoplasia, decreases DNA damage repair, and predicts therapy responses. Oncogene 2023; 42:1857-1873. [PMID: 37095257 PMCID: PMC10238272 DOI: 10.1038/s41388-023-02668-9] [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: 12/28/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/26/2023]
Abstract
Prostate cancer (PCa), the second leading cause of death in American men, includes distinct genetic subtypes with distinct therapeutic vulnerabilities. The DACH1 gene encodes a winged helix/Forkhead DNA-binding protein that competes for binding to FOXM1 sites. Herein, DACH1 gene deletion within the 13q21.31-q21.33 region occurs in up to 18% of human PCa and was associated with increased AR activity and poor prognosis. In prostate OncoMice, prostate-specific deletion of the Dach1 gene enhanced prostatic intraepithelial neoplasia (PIN), and was associated with increased TGFβ activity and DNA damage. Reduced Dach1 increased DNA damage in response to genotoxic stresses. DACH1 was recruited to sites of DNA damage, augmenting recruitment of Ku70/Ku80. Reduced Dach1 expression was associated with increased homology directed repair and resistance to PARP inhibitors and TGFβ kinase inhibitors. Reduced Dach1 expression may define a subclass of PCa that warrants specific therapies.
Collapse
Affiliation(s)
- Zhiping Li
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
- Dxige Research, Courtenay, BC, V9N 1C2, Canada
| | - Gabriele Di Sante
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Anthony W Ashton
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - Agnese DiRocco
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Min Wang
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jun Zhao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Sankar Addya
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Chenguang Wang
- Department of Cancer Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Peter A McCue
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Mt. Sinai, Hospital, 1468 Madison Ave., Floor 15, New York, NY, 10029, USA
| | - Runzhi Liu
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Kishan Patel
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Rasha Hamid
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - Jorim Parmar
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
| | - James B DuHadaway
- Lankenau Institute for Medical Research, 100 East Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, VSZ 4S6, Canada
| | - Mathew C Casimiro
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA
- Abraham Baldwin Agricultural College, Department of Science and Mathematics, Box 15, 2802 Moore Highway, Tifton, GA, 31794, USA
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, 3601 Spruce St., Philadelphia, PA, 19104, USA
| | - Lai Yee Phoon
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Hao Chen
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Li Lan
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Richard G Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA, 18902, USA.
- The Wistar Cancer Center, Philadelphia, PA, 19104, USA.
| |
Collapse
|
44
|
Elliott KH, Balchand SK, Bonatto Paese CL, Chang CF, Yang Y, Brown KM, Rasicci DT, He H, Thorner K, Chaturvedi P, Murray SA, Chen J, Porollo A, Peterson KA, Brugmann SA. Identification of a heterogeneous and dynamic ciliome during embryonic development and cell differentiation. Development 2023; 150:dev201237. [PMID: 36971348 PMCID: PMC10163354 DOI: 10.1242/dev.201237] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Primary cilia are nearly ubiquitous organelles that transduce molecular and mechanical signals. Although the basic structure of the cilium and the cadre of genes that contribute to ciliary formation and function (the ciliome) are believed to be evolutionarily conserved, the presentation of ciliopathies with narrow, tissue-specific phenotypes and distinct molecular readouts suggests that an unappreciated heterogeneity exists within this organelle. Here, we provide a searchable transcriptomic resource for a curated primary ciliome, detailing various subgroups of differentially expressed genes within the ciliome that display tissue and temporal specificity. Genes within the differentially expressed ciliome exhibited a lower level of functional constraint across species, suggesting organism and cell-specific function adaptation. The biological relevance of ciliary heterogeneity was functionally validated by using Cas9 gene-editing to disrupt ciliary genes that displayed dynamic gene expression profiles during osteogenic differentiation of multipotent neural crest cells. Collectively, this novel primary cilia-focused resource will allow researchers to explore longstanding questions related to how tissue and cell-type specific functions and ciliary heterogeneity may contribute to the range of phenotypes associated with ciliopathies.
Collapse
Affiliation(s)
- Kelsey H. Elliott
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | - Sai K. Balchand
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | - Christian Louis Bonatto Paese
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | - Ching-Fang Chang
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | - Yanfen Yang
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | - Kari M. Brown
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
| | | | - Hao He
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Konrad Thorner
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
| | - Praneet Chaturvedi
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
| | | | - Jing Chen
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
| | - Aleksey Porollo
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
| | | | - Samantha A. Brugmann
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical, Cincinnati, OH 45229, USA
- University of Cincinnati, College of Medicine, Department of Pediatrics, Cincinnati, OH 45229, USA
- Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
45
|
Amemiya H, Yamamoto M, Higa K, Watanabe G, Taniguchi S, Kitamura K, Jeong J, Yanagisawa N, Fukuda KI, Abe S. Effects of Myostatin on Nuclear Morphology at the Myotendinous Junction. Int J Mol Sci 2023; 24:ijms24076634. [PMID: 37047606 PMCID: PMC10094852 DOI: 10.3390/ijms24076634] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Myostatin (Myo) is known to suppress skeletal muscle growth, and was recently reported to control tendon homeostasis. The purpose of the present study was to investigate the regulatory involvement of Myo in the myotendinous junction (MTJ) in vivo and in vitro. After Achilles tendon injury in mice, we identified unexpected cell accumulation on the tendon side of the MTJ. At postoperative day 7 (POD7), the nuclei had an egg-like profile, whereas at POD28 they were spindle-shaped. The aspect ratio of nuclei on the tendon side of the MTJ differed significantly between POD7 and POD28 (p = 4.67 × 10−34). We then investigated Myo expression in the injured Achilles tendon. At the MTJ, Myo expression was significantly increased at POD28 relative to POD7 (p = 0.0309). To investigate the action of Myo in vitro, we then prepared laminated sheets of myoblasts (C2C12) and fibroblasts (NIH3T3) (a pseudo MTJ model). Myo did not affect the expression of Pax7 and desmin (markers of muscle development), scleraxis and temonodulin (markers of tendon development), or Sox9 (a common marker of muscle and tendon development) in the cell sheets. However, Myo changed the nuclear morphology of scleraxis-positive cells arrayed at the boundary between the myoblast sheet and the fibroblast sheet (aspect ratio of the cell nuclei, myostatin(+) vs. myostatin(-): p = 0.000134). Myo may strengthen the connection at the MTJ in the initial stages of growth and wound healing.
Collapse
Affiliation(s)
- Hikari Amemiya
- Division of Special Needs Dentistry and Orofacial Pain, Department of Oral Health and Clinical Science, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Masahito Yamamoto
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kazunari Higa
- Ophthalmology/Cornea Center, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa, Chiba 272-8513, Japan
| | - Genji Watanabe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shuichiro Taniguchi
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Kei Kitamura
- Department of Histology and Developmental Biology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, 345 E. 24th Street, New York, NY 10010, USA
| | - Nobuaki Yanagisawa
- Division of Oral Health Sciences, Department of Health Sciences, School of Health and Social Services, Saitama Prefectural University, 820 Sannomia, Koshigaya-shi, Saitama 343-0036, Japan
| | - Ken-ichi Fukuda
- Division of Special Needs Dentistry and Orofacial Pain, Department of Oral Health and Clinical Science, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| | - Shinichi Abe
- Department of Anatomy, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan
| |
Collapse
|
46
|
Colacci A, Corvi R, Ohmori K, Paparella M, Serra S, Da Rocha Carrico I, Vasseur P, Jacobs MN. The Cell Transformation Assay: A Historical Assessment of Current Knowledge of Applications in an Integrated Approach to Testing and Assessment for Non-Genotoxic Carcinogens. Int J Mol Sci 2023; 24:ijms24065659. [PMID: 36982734 PMCID: PMC10057754 DOI: 10.3390/ijms24065659] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
The history of the development of the cell transformation assays (CTAs) is described, providing an overview of in vitro cell transformation from its origin to the new transcriptomic-based CTAs. Application of this knowledge is utilized to address how the different types of CTAs, variously addressing initiation and promotion, can be included on a mechanistic basis within the integrated approach to testing and assessment (IATA) for non-genotoxic carcinogens. Building upon assay assessments targeting the key events in the IATA, we identify how the different CTA models can appropriately fit, following preceding steps in the IATA. The preceding steps are the prescreening transcriptomic approaches, and assessment within the earlier key events of inflammation, immune disruption, mitotic signaling and cell injury. The CTA models address the later key events of (sustained) proliferation and change in morphology leading to tumor formation. The complementary key biomarkers with respect to the precursor key events and respective CTAs are mapped, providing a structured mechanistic approach to represent the complexity of the (non-genotoxic) carcinogenesis process, and specifically their capacity to identify non-genotoxic carcinogenic chemicals in a human relevant IATA.
Collapse
Affiliation(s)
- Annamaria Colacci
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Via Po 5, I-40139 Bologna, Italy
- Correspondence:
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), I-21027 Ispra, Italy
| | - Kyomi Ohmori
- Chemical Division, Kanagawa Prefectural Institute of Public Health, Chigasaki 253-0087, Japan
- Research Initiatives and Promotion Organization, Yokohama National University, Yokohama 240-8501, Japan
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, A-6020 Innbruck, Austria
| | - Stefania Serra
- Agency for Prevention, Environment and Energy, Emilia-Romagna (Arpae), Via Po 5, I-40139 Bologna, Italy
| | | | - Paule Vasseur
- Universite de Lorraine, CNRS UMR 7360 LIEC, Laboratoire Interdisciplinaire des Environnements Continentaux, 57070 Metz, France
| | - Miriam Naomi Jacobs
- Radiation, Chemical and Environmental Hazards, UK Health Security Agency, Harwell Science and Innovation Campus, Chilton OX11 0RQ, UK
| |
Collapse
|
47
|
Pitt K, Mochida Y, Senoo M. Greener Grass: The Modern History of Epithelial Stem Cell Innovation. Life (Basel) 2023; 13:688. [PMID: 36983843 PMCID: PMC10058258 DOI: 10.3390/life13030688] [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: 02/07/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
The field of epithelial stem cell development has been irrevocably shaped by the work of American scientist Howard Green, whose breakthroughs in stem cell culture methods translated to therapeutic practice. In this review, we chronicle the milestones that propelled the field of regenerative medicine of the skin forward over the last fifty years. We detail the early discoveries made by Green and his collaborators, highlight clinical cases that made life-saving use of his findings, and discuss the accomplishments of other scientists who later innovated upon his discoveries.
Collapse
Affiliation(s)
- Keshia Pitt
- Graduate Program in Molecular and Translational Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Yoshiyuki Mochida
- Graduate Program in Molecular and Translational Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Makoto Senoo
- Graduate Program in Molecular and Translational Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, 72 East Concord Street, Boston, MA 02118, USA
- Cell Exosome Therapeutics Inc., 2-16-9 Higashi, Shibuya-ku, Tokyo 150-0011, Japan
| |
Collapse
|
48
|
Perrelli A, Bozza A, Ferraris C, Osella S, Moglia A, Mioletti S, Battaglia L, Retta SF. Multidrug-Loaded Lipid Nanoemulsions for the Combinatorial Treatment of Cerebral Cavernous Malformation Disease. Biomedicines 2023; 11:biomedicines11020480. [PMID: 36831015 PMCID: PMC9953270 DOI: 10.3390/biomedicines11020480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/04/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Cerebral cavernous malformation (CCM) or cavernoma is a major vascular disease of genetic origin, whose main phenotypes occur in the central nervous system, and is currently devoid of pharmacological therapeutic strategies. Cavernomas can remain asymptomatic during a lifetime or manifest with a wide range of symptoms, including recurrent headaches, seizures, strokes, and intracerebral hemorrhages. Loss-of-function mutations in KRIT1/CCM1 are responsible for more than 50% of all familial cases, and have been clearly shown to affect cellular junctions, redox homeostasis, inflammatory responses, and angiogenesis. In this study, we investigated the therapeutic effects of multidrug-loaded lipid nanoemulsions in rescuing the pathological phenotype of CCM disease. The pro-autophagic rapamycin, antioxidant avenanthramide, and antiangiogenic bevacizumab were loaded into nanoemulsions, with the aim of reducing the major molecular dysfunctions associated with cavernomas. Through Western blot analysis of biomarkers in an in vitro CCM model, we demonstrated that drug-loaded lipid nanoemulsions rescue antioxidant responses, reactivate autophagy, and reduce the effect of pro-angiogenic factors better than the free drugs. Our results show the importance of developing a combinatorial preventive and therapeutic approach to reduce the risk of lesion formation and inhibit or completely revert the multiple hallmarks that characterize the pathogenesis and progression of cavernomas.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, Rochester, NY 14620, USA
| | - Annalisa Bozza
- Department of Drug Science and Technology, University of Torino, 10125 Torino, TO, Italy
| | - Chiara Ferraris
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
| | - Sara Osella
- San Giovanni Bosco Hospital, University of Torino, 10154 Torino, TO, Italy
| | - Andrea Moglia
- Department of Agricultural, Forest and Food Sciences, University of Torino, 10095 Grugliasco, TO, Italy
| | - Silvia Mioletti
- Department of Veterinary Sciences, University of Torino, 10095 Grugliasco, TO, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Torino, 10125 Torino, TO, Italy
- Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, 10124 Torino, TO, Italy
- Correspondence: (L.B.); (S.F.R.)
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, TO, Italy
- Correspondence: (L.B.); (S.F.R.)
| |
Collapse
|
49
|
Gonçalves AM, Leal F, Moreira A, Schellhorn T, Blahnová VH, Zeiringer S, Vocetková K, Tetyczka C, Simaite A, Buzgo M, Roblegg E, Costa PF, Ertl P, Filová E, Kohl Y. Potential of Electrospun Fibrous Scaffolds for Intestinal, Skin, and Lung Epithelial Tissue Modeling. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
| | - Filipa Leal
- BIOFABICS Rua Alfredo Allen 455 4200-135 Porto Portugal
| | | | - Tobias Schellhorn
- Institute of Chemical Technologies and Analytics Vienna University of Technology Getreidemarkt 9/164 1060 Vienna Austria
| | - Veronika Hefka Blahnová
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Scarlett Zeiringer
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | - Karolina Vocetková
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Carolin Tetyczka
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | - Aiva Simaite
- InoCure s.r.o. Politických vězňů 935/13 11000 Praha 1 Prague Czech Republic
| | - Matej Buzgo
- BIOFABICS Rua Alfredo Allen 455 4200-135 Porto Portugal
| | - Eva Roblegg
- Institute of Pharmaceutical Sciences University of Graz Universitaetsplatz 1 8010 Graz Austria
| | | | - Peter Ertl
- Institute of Chemical Technologies and Analytics Vienna University of Technology Getreidemarkt 9/164 1060 Vienna Austria
| | - Eva Filová
- Institute of Experimental Medicine of the Czech Academy of Sciences Vídeňská 1083 14220 Prague Czechia
| | - Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT Joseph-von-Fraunhofer-Weg 1 66280 Sulzbach/Saar Germany
| |
Collapse
|
50
|
Vazquez-Sandoval A, Velez-delValle C, Hernández-Mosqueira C, Marsch-Moreno M, Ayala-Sumuano JT, Kuri-Harcuch W. FAM129B is a cooperative protein that regulates adipogenesis. Biochem Biophys Res Commun 2023; 638:66-75. [PMID: 36442234 DOI: 10.1016/j.bbrc.2022.11.042] [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: 10/28/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
FAM129B is one of Niban-like proteins described in neoplastic cells and implicated in melanoma cell invasion, but no reports have been published on FAM129B and cell differentiation. We show that FAM129B is early and transiently expressed and crucial for 3T3-F442A adipogenesis. Fam129b is expressed downstream of the early genes Cebpb, Klf4, Klf5 and Srebf1a, but upstream of Pparg2 since knockdown of Fam129b blocked Pparg2 expression and adipose differentiation. Glycogen synthase kinase 3 beta activity, a crucial kinase for adipogenesis, and the ERK1/2 are involved in FAM129B phosphorylation as part of the adipogenic program. Phosphorylated FAM129B is crucial for Pparg2 expression and the lipogenic gene expression downstream of Pparg2, and hence for adipogenesis. Fam129b knockdown reduced adipocyte cluster formation and size, regulating commitment and clonal amplification. In vivo, BAT, inguinal and epidydimal fat expressed Fam129b, suggesting a role in adipose tissue development. We conclude that FAM129B is a cooperative protein that regulates differentiation during the early stages of adipogenesis.
Collapse
Affiliation(s)
- Alfredo Vazquez-Sandoval
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Cristina Velez-delValle
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Claudia Hernández-Mosqueira
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Meytha Marsch-Moreno
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Jorge-Tonatiuh Ayala-Sumuano
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico; Department of Biomedical Research, IDIX Biotech, Avenida de Los Portones 1151, Queretaro, CP 76100, Mexico
| | - Walid Kuri-Harcuch
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico.
| |
Collapse
|