1
|
Wang W, Tian X, Yan L, Guan X, Dong B, Zhao M, Liu D, Wu J, Hao C. Identification of the γ-glutamyl cycle as a novel therapeutic target and 5-oxoproline as a new biomarker for diagnosing pancreatic cancer. Ann Med 2023; 55:2242247. [PMID: 37544888 PMCID: PMC10405758 DOI: 10.1080/07853890.2023.2242247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/23/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant solid tumours, and abnormal metabolic reprogramming in the tumour microenvironment is regarded as an important contributor to its pathogenesis. OBJECTIVES As there is an urgency to identify new targets based on the metabolic features that are highly refractory to PDAC treatment, this study aimed to identify suitable therapeutic targets for PDAC. METHODS In this study, gene set enrichment and Kyoto Encyclopedia of Genes and Genomes analyses were performed on 163 PDAC tissue samples and 165 normal pancreatic tissue samples from The Cancer Genome Atlas and Genotype-Tissue Expression databases to identify alterations in critical metabolites that may contribute to PDAC pathogenesis. Furthermore, ultra-performance liquid chromatography-tandem mass spectrometry was performed to identify significant metabolic pathways between 24 pairs of tumour and adjacent non-tumour tissues and between serum samples from PDAC patients and healthy donors. RESULTS Fifty-one tissue metabolites and 26 serum metabolites were altered in PDAC. Among them, those in the γ-glutamyl cycle were the most substantially changed, and 5-oxoproline was the biomarker of PDAC with the most significantly decreased levels. CONCLUSIONS The γ-glutamyl cycle and 5-oxoproline might be potential biomarkers and therapeutic targets to improve the diagnosis, therapy, and prognosis of PDAC.
Collapse
Affiliation(s)
- Weijia Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
- Department of International Medical Services (IMS), Beijing Tiantan Hospital of Capital Medical University, Beijing, People’s Republic of China
| | - Xiuyun Tian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Liang Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xiaoya Guan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Bin Dong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Min Zhao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Daoning Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Jianhui Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Chunyi Hao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery/Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Manetti F, Maresca L, Crivaro E, Pepe S, Cini E, Singh S, Governa P, Maramai S, Giannini G, Stecca B, Petricci E. Quinolines and Oxazino-quinoline Derivatives as Small Molecule GLI1 Inhibitors Identified by Virtual Screening. ACS Med Chem Lett 2022; 13:1329-1336. [PMID: 35978701 PMCID: PMC9377010 DOI: 10.1021/acsmedchemlett.2c00249] [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: 05/26/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
A virtual screening approach based on a five-feature pharmacophoric model for negative modulators of GLI1 was applied to databases of commercially available compounds. The resulting quinoline derivatives showed significant ability to reduce the GLI1 protein level and were characterized by submicromolar antiproliferative activity toward human melanoma A375 and medulloblastoma DAOY cell lines. Decoration of the quinoline ring and chemical rigidification to an oxazino-quinoline scaffold allowed us to deduce SAR considerations for future ligand optimization.
Collapse
Affiliation(s)
- Fabrizio Manetti
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| | - Luisa Maresca
- Istituto
per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Viale Pieraccini 6, I-50139 Firenze, Italy
| | - Enrica Crivaro
- Istituto
per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Viale Pieraccini 6, I-50139 Firenze, Italy
| | - Sara Pepe
- Istituto
per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Viale Pieraccini 6, I-50139 Firenze, Italy
| | - Elena Cini
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| | - Snigdha Singh
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| | - Paolo Governa
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| | - Samuele Maramai
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| | | | - Barbara Stecca
- Istituto
per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Viale Pieraccini 6, I-50139 Firenze, Italy
| | - Elena Petricci
- Dipartimento
di Biotecnologie Chimica e Farmacia, Università
di Siena, via A. Moro 2, I-53100 Siena, Italy
| |
Collapse
|
3
|
Maimó-Barceló A, Martín-Saiz L, Fernández JA, Pérez-Romero K, Garfias-Arjona S, Lara-Almúnia M, Piérola-Lopetegui J, Bestard-Escalas J, Barceló-Coblijn G. Polyunsaturated Fatty Acid-Enriched Lipid Fingerprint of Glioblastoma Proliferative Regions Is Differentially Regulated According to Glioblastoma Molecular Subtype. Int J Mol Sci 2022; 23:ijms23062949. [PMID: 35328369 PMCID: PMC8949316 DOI: 10.3390/ijms23062949] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) represents one of the deadliest tumors owing to a lack of effective treatments. The adverse outcomes are worsened by high rates of treatment discontinuation, caused by the severe side effects of temozolomide (TMZ), the reference treatment. Therefore, understanding TMZ’s effects on GBM and healthy brain tissue could reveal new approaches to address chemotherapy side effects. In this context, we have previously demonstrated the membrane lipidome is highly cell type-specific and very sensitive to pathophysiological states. However, little remains known as to how membrane lipids participate in GBM onset and progression. Hence, we employed an ex vivo model to assess the impact of TMZ treatment on healthy and GBM lipidome, which was established through imaging mass spectrometry techniques. This approach revealed that bioactive lipid metabolic hubs (phosphatidylinositol and phosphatidylethanolamine plasmalogen species) were altered in healthy brain tissue treated with TMZ. To better understand these changes, we interrogated RNA expression and DNA methylation datasets of the Cancer Genome Atlas database. The results enabled GBM subtypes and patient survival to be linked with the expression of enzymes accounting for the observed lipidome, thus proving that exploring the lipid changes could reveal promising therapeutic approaches for GBM, and ways to ameliorate TMZ side effects.
Collapse
Affiliation(s)
- Albert Maimó-Barceló
- Institut d’Investigacio Sanitaria Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), 07120 Palma, Spain; (A.M.-B.); (K.P.-R.); (J.P.-L.)
- Research Unit, University Hospital Son Espases, 07120 Palma, Spain
| | - Lucía Martín-Saiz
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain; (L.M.-S.); (J.A.F.)
| | - José A. Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain; (L.M.-S.); (J.A.F.)
| | - Karim Pérez-Romero
- Institut d’Investigacio Sanitaria Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), 07120 Palma, Spain; (A.M.-B.); (K.P.-R.); (J.P.-L.)
- Research Unit, University Hospital Son Espases, 07120 Palma, Spain
| | - Santiago Garfias-Arjona
- Quirónsalud Medical Center, 07300 Inca, Spain;
- Son Verí Quirónsalud Hospital, Balearic Islands, 07609 Son Veri Nou, Spain
- Hospital de Llevant, 07680 Porto Cristo, Spain
| | - Mónica Lara-Almúnia
- Department of Neurosurgery, Jimenez Diaz Foundation University Hospital, Reyes Catolicos Av., No 2, 28040 Madrid, Spain;
- Ruber International Hospital, Maso St., No 38, 28034 Madrid, Spain
| | - Javier Piérola-Lopetegui
- Institut d’Investigacio Sanitaria Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), 07120 Palma, Spain; (A.M.-B.); (K.P.-R.); (J.P.-L.)
- Research Unit, University Hospital Son Espases, 07120 Palma, Spain
| | - Joan Bestard-Escalas
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Bruxelles, Belgium
- Correspondence: (J.B.-E.); (G.B.-C.)
| | - Gwendolyn Barceló-Coblijn
- Institut d’Investigacio Sanitaria Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), 07120 Palma, Spain; (A.M.-B.); (K.P.-R.); (J.P.-L.)
- Research Unit, University Hospital Son Espases, 07120 Palma, Spain
- Correspondence: (J.B.-E.); (G.B.-C.)
| |
Collapse
|
4
|
Garay MI, Comba A, Vara Messler M, Barotto NN, Silva RA, Repossi G, Quiroga PL, Mazzudulli GM, Brunotto MN, Pasqualini ME. Tumorigenic effect mediated by ROS/eicosanoids and their regulation on TP53 expression in a murine mammary gland adenocarcinoma. Prostaglandins Other Lipid Mediat 2021; 155:106564. [PMID: 34004336 DOI: 10.1016/j.prostaglandins.2021.106564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 03/23/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
The aim of this study was to investigate the in vivo and in vitro effects of dietary ω-6 and ω-3 polyunsaturated fatty acids (PUFAs) and their derivatives on the expression of TP53 and their relationship with cellular proliferation and death in a murine mammary adenocarcinoma model. BALB/c mice were divided in three diet groups: chia oil (ChO) rich in ω-3, corn oil (CO) rich in ω-6/ω-3 and safflower oil (SO) rich in ω-6 and subcutaneously inoculated with LMM3 mammary tumor cell line. Results demonstrated that diets with higher concentration of omega-6 PUFAs induced an increment of linoleic and arachidonic acid on tumor cell membranes increasing ROS liberation, 12(S)-HHT generation, TP53, Ki67 expression and cell proliferation. However, diets enriched with high content in omega-3 PUFAs induced higher tumor cell apoptosis and tumor infiltration of CD3+ lymphocytes, lowest cell viability and proliferation. Dietary omega-3 PUFAs nutritional intervention can be used as a potential preventative strategy to inhibit the molecular signaling pathways involved in the mammary tumor growth process as the TP53.
Collapse
Affiliation(s)
- M I Garay
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Ciudad Universitaria, 5000 Córdoba, Argentina; Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - A Comba
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Ciudad Universitaria, 5000 Córdoba, Argentina; Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, 48109, MI, USA.
| | - M Vara Messler
- Dipartimento di Oncologia, Università di Torino, 10124 Torino, Italy.
| | - N N Barotto
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - R A Silva
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - G Repossi
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Ciudad Universitaria, 5000 Córdoba, Argentina; Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - P L Quiroga
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - G M Mazzudulli
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - M N Brunotto
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| | - M E Pasqualini
- Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Ciudad Universitaria, 5000 Córdoba, Argentina; Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina; Instituto de Biología Celular, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Ciudad Universitaria, 5000 Córdoba, Argentina.
| |
Collapse
|
5
|
Avery JT, Zhang R, Boohaker RJ. GLI1: A Therapeutic Target for Cancer. Front Oncol 2021; 11:673154. [PMID: 34113570 PMCID: PMC8186314 DOI: 10.3389/fonc.2021.673154] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
GLI1 is a transcriptional effector at the terminal end of the Hedgehog signaling (Hh) pathway and is tightly regulated during embryonic development and tissue patterning/differentiation. GLI1 has low-level expression in differentiated tissues, however, in certain cancers, aberrant activation of GLI1 has been linked to the promotion of numerous hallmarks of cancer, such as proliferation, survival, angiogenesis, metastasis, metabolic rewiring, and chemotherapeutic resistance. All of these are driven, in part, by GLI1’s role in regulating cell cycle, DNA replication and DNA damage repair processes. The consequences of GLI1 oncogenic activity, specifically the activity surrounding DNA damage repair proteins, such as NBS1, and cell cycle proteins, such as CDK1, can be linked to tumorigenesis and chemoresistance. Therefore, understanding the underlying mechanisms driving GLI1 dysregulation can provide prognostic and diagnostic biomarkers to identify a patient population that would derive therapeutic benefit from either direct inhibition of GLI1 or targeted therapy towards proteins downstream of GLI1 regulation.
Collapse
Affiliation(s)
- Justin T Avery
- Oncology Department, Drug Discovery Division, Southern Research, Birmingham, AL, United States
| | - Ruowen Zhang
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Rebecca J Boohaker
- Oncology Department, Drug Discovery Division, Southern Research, Birmingham, AL, United States
| |
Collapse
|
6
|
Carr RM, Vorobyev D, Lasho T, Marks DL, Tolosa EJ, Vedder A, Almada LL, Yurcheko A, Padioleau I, Alver B, Coltro G, Binder M, Safgren SL, Horn I, You X, Solary E, Balasis ME, Berger K, Hiebert J, Witzig T, Buradkar A, Graf T, Valent P, Mangaonkar AA, Robertson KD, Howard MT, Kaufmann SH, Pin C, Fernandez-Zapico ME, Geissler K, Droin N, Padron E, Zhang J, Nikolaev S, Patnaik MM. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis. Nat Commun 2021; 12:2901. [PMID: 34006870 PMCID: PMC8131698 DOI: 10.1038/s41467-021-23186-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Proliferative chronic myelomonocytic leukemia (pCMML), an aggressive CMML subtype, is associated with dismal outcomes. RAS pathway mutations, mainly NRASG12D, define the pCMML phenotype as demonstrated by our exome sequencing, progenitor colony assays and a Vav-Cre-NrasG12D mouse model. Further, these mutations promote CMML transformation to acute myeloid leukemia. Using a multiomics platform and biochemical and molecular studies we show that in pCMML RAS pathway mutations are associated with a unique gene expression profile enriched in mitotic kinases such as polo-like kinase 1 (PLK1). PLK1 transcript levels are shown to be regulated by an unmutated lysine methyl-transferase (KMT2A) resulting in increased promoter monomethylation of lysine 4 of histone 3. Pharmacologic inhibition of PLK1 in RAS mutant patient-derived xenografts, demonstrates the utility of personalized biomarker-driven therapeutics in pCMML.
Collapse
MESH Headings
- Animals
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Gene Expression Profiling/methods
- Gene Expression Regulation, Leukemic
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Kaplan-Meier Estimate
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Leukemia, Myelomonocytic, Chronic/therapy
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Mutation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Signal Transduction/genetics
- Stem Cell Transplantation/methods
- Transplantation, Homologous
- Exome Sequencing/methods
- Xenograft Model Antitumor Assays/methods
- Polo-Like Kinase 1
- Mice
Collapse
Affiliation(s)
- Ryan M Carr
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Denis Vorobyev
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France
| | - Terra Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - David L Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Ezequiel J Tolosa
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Alexis Vedder
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Andrey Yurcheko
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Bonnie Alver
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, MN, USA
| | - Giacomo Coltro
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Moritz Binder
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Stephanie L Safgren
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Isaac Horn
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, MN, USA
| | - Xiaona You
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric Solary
- INSERM U1170 and Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Maria E Balasis
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Kurt Berger
- London Regional Transgenic and Gene Targeting Facility, Lawson Health Research Institute University of Western Ontario, London, ON, Canada
| | - James Hiebert
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Thomas Witzig
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Ajinkya Buradkar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Temeida Graf
- 5TH Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- 5TH Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | - Keith D Robertson
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, MN, USA
| | - Matthew T Howard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, MN, USA
| | - Scott H Kaufmann
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA
| | - Christopher Pin
- London Regional Transgenic and Gene Targeting Facility, Lawson Health Research Institute University of Western Ontario, London, ON, Canada
| | | | | | - Nathalie Droin
- INSERM U1170 and Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Eric Padron
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Sergey Nikolaev
- INSERM U981, Gustave Roussy Cancer Center, Villejuif, France.
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN, USA.
| |
Collapse
|
7
|
GLI1/GLI2 functional interplay is required to control Hedgehog/GLI targets gene expression. Biochem J 2021; 477:3131-3145. [PMID: 32766732 DOI: 10.1042/bcj20200335] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023]
Abstract
The Hedgehog-regulated transcription factors GLI1 and GLI2 play overlapping roles in development and disease; however, the mechanisms underlying their interplay remain elusive. We report for the first time that GLI1 and GLI2 physically and functionally interact in cancer cells. GLI1 and GLI2 were shown to co-immunoprecipitate in PANC1 pancreatic cancer cells and RMS13 rhabdomyosarcoma cells. Mapping analysis demonstrated that the zinc finger domains of both proteins are required for their heteromerization. RNAi knockdown of either GLI1 or GLI2 inhibited expression of many well-characterized GLI target genes (BCL2, MYCN, PTCH2, IL7 and CCND1) in PANC1 cells, whereas PTCH1 expression was only inhibited by GLI1 depletion. qPCR screening of a large set of putative canonical and non-canonical Hedgehog/GLI targets identified further genes (e.g. E2F1, BMP1, CDK2) strongly down-regulated by GLI1 and/or GLI2 depletion in PANC1 cells, and demonstrated that ANO1, AQP1 and SOCS1 are up-regulated by knockdown of either GLI1 or GLI2. Chromatin immunoprecipitation showed that GLI1 and GLI2 occupied the same regions at the BCL2, MYCN and CCND1 promoters. Furthermore, depletion of GLI1 inhibited GLI2 occupancy at these promoters, suggesting that GLI1/GLI2 interaction is required for the recruitment of GLI2 to these sites. Together, these findings indicate that GLI1 and GLI2 co-ordinately regulate the transcription of some genes, and provide mechanistic insight into the roles of GLI proteins in carcinogenesis.
Collapse
|
8
|
Tsuji S, Ohno Y, Nakamura S, Yamada T, Noda Y, Saio M, Iwama T, Shimazawa M, Hara H. Temozolomide has anti-tumor effects through the phosphorylation of cPLA 2 on glioblastoma cells. Brain Res 2019; 1723:146396. [PMID: 31442413 DOI: 10.1016/j.brainres.2019.146396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 11/18/2022]
Abstract
Temozolomide is an alkylating agent used as the first line of treatment for glioblastoma. However, chemoresistance to temozolomide is common in glioma patients. In addition, there are likely many unknown mechanisms for the anti-tumor effects of temozolomide. It is known that an alkylating agent, sulfur mustard, activates cytosolic phospholipase A2 (cPLA₂) releasing arachidonic acid to suppress tumors. The present study was performed to elucidate the involvement of cPLA2 in the anti-tumor mechanisms of temozolomide. In three glioblastoma cell lines (GL261, U251MG and T98G), we performed several evaluations including cell viability, cell migration and apoptosis, to study temozolomide-induced anti-tumor effects. Further, we evaluated tumor size in the murine orthotropic glioblastoma model after oral administration of temozolomide. Finally, we investigated the phosphorylation of cPLA2 in GL261 cells treated with temozolomide, and clarified whether phosphorylation of cPLA2 affects cell growth. Temozolomide suppressed cell growth and cell migration in glioblastoma cells in vitro and showed anti-tumor effect in the murine orthotopic glioblastoma model in vivo. Furthermore, temozolomide increased phosphorylation of cPLA2, which was associated with suppression of cell growth. However, in MGMT high-expressing glioblastoma T98G cells, temozolomide could not suppress cell growth or cause phosphorylation of cPLA2. These findings indicate that temozolomide suppressed cell growth partly by phosphorylation of cPLA2 in glioblastoma cells. In addition, because temozolomide did not cause phosphorylation of cPLA2 in MGMT high-expressing glioblastoma T98G cells, phosphorylation of cPLA2 may be caused by DNA alkylation of temozolomide.
Collapse
Affiliation(s)
- Shohei Tsuji
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Yuta Ohno
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Gifu 501-0023, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Tetsuya Yamada
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan; Department of Neurosurgery, Gifu University School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Yasuhiro Noda
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Masanao Saio
- Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan.
| |
Collapse
|
9
|
Xu S, Shu P, Zou S, Shen X, Qu Y, Zhang Y, Sun K, Zhang J. NFATc1 is a tumor suppressor in hepatocellular carcinoma and induces tumor cell apoptosis by activating the FasL-mediated extrinsic signaling pathway. Cancer Med 2018; 7:4701-4717. [PMID: 30085405 PMCID: PMC6143940 DOI: 10.1002/cam4.1716] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 07/07/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a family of transcription factors that have important functions in many tumors. However, the expression level and functional role of NFAT in hepatocellular carcinoma (HCC) remain unclear. In this study, we showed that NFATc1 expression was decreased in both HCC tissues and cell lines. Low expression of NFATc1 was correlated with larger tumor size, advanced tumor‐node‐metastasis (TNM) stage, high serum AFP level, and liver cirrhosis. Furthermore, patients with low NFATc1 expression exhibited poor prognosis. Ectopic expression of NFATc1 in HCC cells inhibited proliferation and colony formation, leading to G1 arrest and induction of apoptosis. In addition, we demonstrated that NFATc1 increased Fas ligand (FasL) expression by directly binding to its promoter and activated the extrinsic apoptotic pathway. NFATc1 and FasL expression patterns and their prognostic value for patients with HCC were also evaluated in TCGA Liver Hepatocellular Carcinoma database. Knock‐down of FasL expression by siRNA in HCC cell lines abolished NFATc1's antiproliferative and pro‐apoptotic effects. In conclusion, NFATc1 is frequently inactivated in HCC and functions as a tumor suppressor in liver carcinogenesis. Ectopic expression of NFATc1 in HCC cells induces apoptosis by activating the FasL‐mediated extrinsic signaling pathway.
Collapse
Affiliation(s)
- Sanrong Xu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Penghao Shu
- Department of Hepatobiliary Surgery, People's Hospital of Danyang, Danyang, China
| | - Song Zou
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaofeng Shen
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuanqian Qu
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yong Zhang
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kang Sun
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jin Zhang
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Comba A, Bonnet LV, Goitea VE, Hallak ME, Galiano MR. Arginylated Calreticulin Increases Apoptotic Response Induced by Bortezomib in Glioma Cells. Mol Neurobiol 2018; 56:1653-1664. [PMID: 29916141 DOI: 10.1007/s12035-018-1182-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/07/2018] [Indexed: 10/14/2022]
Abstract
After retrotranslocation from the endoplasmic reticulum to the cytoplasm, calreticulin is modified by the enzyme arginyltransferase-1 (ATE1). Cellular levels of arginylated calreticulin (R-CRT) are regulated in part by the proteasomal system. Under various stress conditions, R-CRT becomes associated with stress granules (SGs) or reaches the plasma membrane (PM), where it participates in pro-apoptotic signaling. The mechanisms underlying the resistance of tumor cells to apoptosis induced by specific drugs remain unclear. We evaluated the regulatory role of R-CRT in apoptosis of human glioma cell lines treated with the proteasome inhibitor bortezomib (BT). Two cell lines (HOG, MO59K) displaying distinctive susceptibility to apoptosis induction were studied further. BT efficiency was found to be correlated with a subcellular distribution of R-CRT. In MO59K (apoptosis-resistant), R-CRT was confined to SGs formed following BT treatment. In contrast, HOG (apoptosis-susceptible) treated with BT showed lower SG formation and higher levels of cytosolic and PM R-CRT. Increased R-CRT level was associated with enhanced mobilization of intracellular Ca2+ and with sustained apoptosis activation via upregulation of cell death receptor DR5. R-CRT overexpression in the cytoplasm of MO59K rendered the cells susceptible to BT-induced, DR5-mediated cell death. Our findings suggest that R-CRT plays an essential role in the effect of BT treatment on tumor cells and that ATE1 is a strong candidate target for future studies of cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Andrea Comba
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Laura V Bonnet
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Victor E Goitea
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Marta E Hallak
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Mauricio R Galiano
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC-CONICET), Universidad Nacional de Córdoba, 5000, Córdoba, Argentina.
| |
Collapse
|
11
|
Mastrangelo E, Milani M. Role and inhibition of GLI1 protein in cancer. LUNG CANCER-TARGETS AND THERAPY 2018; 9:35-43. [PMID: 29628779 PMCID: PMC5877502 DOI: 10.2147/lctt.s124483] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
GLI1 is a transcriptional regulator involved in the development of different types of cancer. GLI1 transcriptional activity is regulated within the Hedgehog pathway (canonical activity), but can also be controlled independently (non-canonical activity) in the context of other signaling pathways. Experimental evidences show GLI1 involvement in both small- and non–small-cell lung cancers. Direct inhibition of the protein, in combination with other chemotherapeutic agents, represents a promising strategy for the treatment of different malignancies.
Collapse
Affiliation(s)
- Eloise Mastrangelo
- CNR - Biophysics Institute, c/o Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Mario Milani
- CNR - Biophysics Institute, c/o Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
12
|
Scherma ME, Madzzuduli G, Silva RA, Garay MI, Repossi G, Brunotto M, Pasqualini ME. The effects of ω-6 and ω-3 fatty-acids on early stages of mice DMBA submandibular glands tumorigenesis. Prostaglandins Leukot Essent Fatty Acids 2017; 125:48-55. [PMID: 28987722 DOI: 10.1016/j.plefa.2017.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/16/2017] [Accepted: 08/21/2017] [Indexed: 01/28/2023]
Abstract
The aim of this work was: to assess the impact of diets enriched in polyunsaturated fatty acids ω-3 and ω-6 families on the lipid profile of cell membrane and their effect on cycle regulation and apoptosis, evaluated by TP53 and Ki-67 expression in 9,10-dimethyl-1,2-benzanthracene (DMBA) induced tumor development in submandibular glands (SMG) in murine models. To generate tumorigenic changes, SMG mice in the experimental group were injected with 50μl of 0.5% of DMBA. Both control (no DMBA) and experimental groups of BALB/c mice were fed with: chia oil (ChO), rich in ω-3 fatty acid; corn oil (CO), rich in ω-6/ω-3 fatty acid; and safflower (SO) oil, rich in ω-6fatty acid. Results demonstrate novel differential effects of ω-3 and ω-6 PUFAs on the regulation of early tumorigenesis events in murine SMG injected with DMBA. This knowledge may help to develop chemoprotective treatments, therapeutic agents and health promotion and prevention activities in humans.
Collapse
Affiliation(s)
- M E Scherma
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Argentina
| | - G Madzzuduli
- Instituto de Investigaciones en Ciencias de la Salud, (INICSA-CONICET-UNC), Argentina
| | - R A Silva
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - M I Garay
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, (INICSA-CONICET-UNC), Argentina
| | - G Repossi
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, (INICSA-CONICET-UNC), Argentina
| | - M Brunotto
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Argentina
| | - M E Pasqualini
- Cátedra de Biología Celular, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, (INICSA-CONICET-UNC), Argentina.
| |
Collapse
|
13
|
Gundala NKV, Naidu VGM, Das UN. Arachidonic acid and lipoxin A4 attenuate alloxan-induced cytotoxicity to RIN5F cells in vitro and type 1 diabetes mellitus in vivo. Biofactors 2017; 43:251-271. [PMID: 27862450 DOI: 10.1002/biof.1336] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/17/2016] [Accepted: 10/03/2016] [Indexed: 12/27/2022]
Abstract
OBJECTIVE We studied whether polyunsaturated fatty acids (PUFAs) can protect rat insulinoma (RIN5F) cells against alloxan-induced apoptosis in vitro and type 1 diabetes mellitus (type 1 DM) in vivo and if so, mechanism of this beneficial action. MATERIAL AND METHODS In vitro study was conducted using RIN5F cells while in vivo study was performed in Wistar rats. The effect of PUFAs, cyclo-oxygenase and lipoxygenase inhibitors, various eicosanoids and PUFAs metabolites: lipoxin A4 (LXA4), resolvin D2 and protectin against alloxan-induced cytotoxicity to RIN5F cells and type 1 DM was studied. Expression of PDX1, P65 NF-kB and IKB in RIN5F cells and Nrf2, GLUT2, COX2, iNOS protein levels in the pancreatic tissue and plasma glucose, insulin and tumor necrosis factor-α and antioxidants, lipid peroxides and nitric oxide were measured. RESULTS Of all, arachidonic acid (AA) was found to be the most effective against alloxan-induced cytotoxicity to RIN5F cells and preventing type 1 DM. Both cyclo-oxygenase and lipoxygenase inhibitors did not block the beneficial actions of AA in vitro and in vivo. Alloxan inhibited LXA4 production by RIN5F cells and in alloxan-induced type 1 DM Wistar rats. AA-treatment restored LXA4 levels to normal both in vitro and in vivo. LXA4 protected RIN5F cells against alloxan-induced cytotoxicity and prevented type 1 DM and restored expression of Nrf2, Glut2, COX2, and iNOS genes and abnormal antioxidants to near normal. DISCUSSION AA seems to bring about its beneficial actions against alloxan-induced cytotoxicity and type 1 DM by enhancing the production of LXA4. © 2016 BioFactors, 43(2):251-271, 2017.
Collapse
Affiliation(s)
- Naveen K V Gundala
- Department of Medicine, BioScience Research Centre, Gayatri Vidya Parishad Hospital, GVP College of Engineering Campus, Visakhapatnam, 530048, India
| | - Vegi G M Naidu
- National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Undurti N Das
- Department of Medicine, BioScience Research Centre, Gayatri Vidya Parishad Hospital, GVP College of Engineering Campus, Visakhapatnam, 530048, India
- UND Life Sciences, 2020 S 360th St, # K-202, Federal Way, WA, 98003, USA
| |
Collapse
|
14
|
Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis 2016; 7:e2199. [PMID: 27100893 PMCID: PMC4855676 DOI: 10.1038/cddis.2016.97] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors consists of four Ca2+-regulated members (NFAT1–NFAT4), which were first described in T lymphocytes. In addition to their well-documented role in T lymphocytes, where they control gene expression during cell activation and differentiation, NFAT proteins are also expressed in a wide range of cells and tissue types and regulate genes involved in cell cycle, apoptosis, angiogenesis and metastasis. The NFAT proteins share a highly conserved DNA-binding domain (DBD), which allows all NFAT members to bind to the same DNA sequence in enhancers or promoter regions. The same DNA-binding specificity suggests redundant roles for the NFAT proteins, which is true during the regulation of some genes such as IL-2 and p21. However, it has become increasingly clear that different NFAT proteins and even isoforms can have unique functions. In this review, we address the possible reasons for these distinct roles, particularly regarding N- and C-terminal transactivation regions (TADs) and the partner proteins that interact with these TADs. We also discuss the genes regulated by NFAT during cell cycle regulation and apoptosis and the role of NFAT during tumorigenesis.
Collapse
|