1
|
Liu W, Cao H, Wang J, Elmusrati A, Han B, Chen W, Zhou P, Li X, Keysar S, Jimeno A, Wang CY. Histone-methyltransferase KMT2D deficiency impairs the Fanconi anemia/BRCA pathway upon glycolytic inhibition in squamous cell carcinoma. Nat Commun 2024; 15:6755. [PMID: 39117659 PMCID: PMC11310337 DOI: 10.1038/s41467-024-50861-5] [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: 12/06/2023] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Histone lysine methyltransferase 2D (KMT2D) is the most frequently mutated epigenetic modifier in head and neck squamous cell carcinoma (HNSCC). However, the role of KMT2D in HNSCC tumorigenesis and whether its mutations confer any therapeutic vulnerabilities remain unknown. Here we show that KMT2D deficiency promotes HNSCC growth through increasing glycolysis. Additionally, KMT2D loss decreases the expression of Fanconi Anemia (FA)/BRCA pathway genes under glycolytic inhibition. Mechanistically, glycolytic inhibition facilitates the occupancy of KMT2D to the promoter/enhancer regions of FA genes. KMT2D loss reprograms the epigenomic landscapes of FA genes by transiting their promoter/enhancer states from active to inactive under glycolytic inhibition. Therefore, combining the glycolysis inhibitor 2-DG with DNA crosslinking agents or poly (ADP-ribose) polymerase (PARP) inhibitors preferentially inhibits tumor growth of KMT2D-deficient mouse HNSCC and patient-derived xenografts (PDXs) harboring KMT2D-inactivating mutations. These findings provide an epigenomic basis for developing targeted therapies for HNSCC patients with KMT2D-inactivating mutations.
Collapse
Affiliation(s)
- Wei Liu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hongchao Cao
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jing Wang
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Areeg Elmusrati
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bing Han
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wei Chen
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ping Zhou
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiyao Li
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephen Keysar
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cun-Yu Wang
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
- Laboratory of Molecular Signaling, Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Wu CJ, Livak F, Ashwell JD. The histone methyltransferase KMT2D maintains cellular glucocorticoid responsiveness by shielding the glucocorticoid receptor from degradation. J Biol Chem 2024; 300:107581. [PMID: 39025450 PMCID: PMC11350265 DOI: 10.1016/j.jbc.2024.107581] [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: 04/01/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
Because of their ability to induce lymphocyte apoptosis, glucocorticoids (GC) are widely used to treat hematological malignancies such as lymphomas and multiple myeloma. Their effectiveness is often limited, however, due to the development of glucocorticoid resistance by a variety of molecular mechanisms. Here we performed an unbiased genome-wide CRISPR screen with the human T-cell leukemia cell line Jurkat to find previously unidentified genes required for GC-induced apoptosis. One such gene was KMT2D (also known as MLL2 or MLL4), which encodes a histone lysine methyltransferase whose mutations are associated with a variety of cancers, blood malignancies in particular, and are considered markers of poor prognosis. Knockout of KMT2D by CRISPR/Cas9 gene editing in Jurkat and several multiple myeloma cell lines downregulated GR protein expression. Surprisingly, this was not due to a reduction in GR transcripts, but rather to a decrease in the protein's half-life, primarily due to proteasomal degradation. Reconstitution of KMT2D expression restored GR levels. In contrast to the known ability of KMT2D to control gene transcription through covalent histone methylation, KMT2D-mediated upregulation of GR levels did not require its methyltransferase activity. Co-immunoprecipitation and proximity ligation assays found constitutive binding of KMT2D to the GR, which was enhanced in the presence of GC. These observations reveal KMT2D to be essential for the stabilization of cellular GR levels, and suggest a possible mechanism by which KMT2D mutations may lead to GC resistance in some malignancies.
Collapse
Affiliation(s)
- Chuan-Jin Wu
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ferenc Livak
- Laboratory of Genome Integrity Flow Cytometry Core, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
3
|
Tompa M, Galik B, Urban P, Kajtar BI, Kraboth Z, Gyenesei A, Miseta A, Kalman B. On the Boundary of Exploratory Genomics and Translation in Sequential Glioblastoma. Int J Mol Sci 2024; 25:7564. [PMID: 39062807 PMCID: PMC11277311 DOI: 10.3390/ijms25147564] [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: 06/11/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
OMICS methods brought significant advancements to the understanding of tumor cell biology, which transformed the treatment and prognosis of several cancers. Clinical practice and outcomes, however, changed significantly less in the case of glioblastoma (GBM). In this study, we aimed to assess the utility of whole exome (WES) sequencing in the clinical setting. Ten pairs of formalin-fixed, paraffin-embedded (FFPE) GBM specimens were obtained at onset (GBM-P) and at recurrence (GBM-R). Histopathological and molecular features of all samples supported the diagnosis of GBM based on WHO CNS5. WES data were filtered, applying a strict and custom-made pipeline, and occurrence of oncogenic and likely oncogenic variants in GBM-P, GBM-R or both were identified by using the VarSeq program version 2.5.0 (Golden Helix, Inc.). Characteristics and recurrence of the variants were analyzed in our own cohort and were also compared to those available in the COSMIC database. The lists of oncogenic and likely oncogenic variants corresponded to those identified in other studies. The average number of these variants were 4 and 5 out of all detected 24 and 34 variants in GBM-P and GBM-R samples, respectively. On average, one shared oncogenic/likely oncogenic variant was found in the pairs. We assessed the identified variants' therapeutic significance, also taking into consideration the guidelines by the Association for Molecular Pathology (AMP). Our data support that a thorough WES analysis is suitable for identifying oncogenic and likely oncogenic variants in an individual clinical sample or a small cohort of FFPE glioma specimens, which concur with those of comprehensive research studies. Such analyses also allow us to monitor molecular dynamics of sequential GBM. In addition, careful evaluation of data according to the AMP guideline reveal that though therapeutic applicability of the variants is generally limited in the clinic, such information may be valuable in selected cases, and can support innovative preclinical and clinical trials.
Collapse
Affiliation(s)
- Marton Tompa
- Szentagothai Research Center, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary; (B.G.); (P.U.); (A.G.)
- Department of Molecular Medicine, Markusovszky University Teaching Hospital, 5. Markusovszky Street, 9700 Szombathely, Hungary
| | - Bence Galik
- Szentagothai Research Center, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary; (B.G.); (P.U.); (A.G.)
| | - Peter Urban
- Szentagothai Research Center, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary; (B.G.); (P.U.); (A.G.)
| | - Bela Istvan Kajtar
- Department of Pathology, School of Medicine, University of Pecs, 12. Szigeti Street, 7624 Pecs, Hungary; (B.I.K.); (Z.K.)
| | - Zoltan Kraboth
- Department of Pathology, School of Medicine, University of Pecs, 12. Szigeti Street, 7624 Pecs, Hungary; (B.I.K.); (Z.K.)
| | - Attila Gyenesei
- Szentagothai Research Center, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary; (B.G.); (P.U.); (A.G.)
| | - Attila Miseta
- Office of the Dean, School of Medicine, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary;
| | - Bernadette Kalman
- Szentagothai Research Center, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary; (B.G.); (P.U.); (A.G.)
- Department of Molecular Medicine, Markusovszky University Teaching Hospital, 5. Markusovszky Street, 9700 Szombathely, Hungary
- Office of the Dean, School of Medicine, University of Pecs, 20. Ifjusag Street, 7624 Pecs, Hungary;
| |
Collapse
|
4
|
Kisor KP, Ruiz DG, Jacobson MP, Barber DL. A role for pH dynamics regulating transcription factor DNA binding selectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595212. [PMID: 38826444 PMCID: PMC11142074 DOI: 10.1101/2024.05.21.595212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Intracellular pH (pHi) dynamics regulates diverse cell processes such as proliferation, dysplasia, and differentiation, often mediated by the protonation state of a functionally critical histidine residue in endogenous pH sensing proteins. How pHi dynamics can directly regulate gene expression and whether transcription factors can function as pH sensors has received limited attention. We tested the prediction that transcription factors with a histidine in their DNA binding domain (DBD) that forms hydrogen bonds with nucleotides can have pH-regulated activity, which is relevant to more than 85 transcription factors in distinct families, including FOX, KLF, SOX and MITF/Myc. Focusing on FOX family transcription factors, we used unbiased SELEX-seq to identify pH-dependent DNA binding motif preferences, then confirm pH-regulated binding affinities for FOXC2, FOXM1, and FOXN1 to a canonical FkhP DNA motif that are 2.5 to 7.5 greater at pH 7.0 compared with pH 7.5. For FOXC2, we also find greater activity for an FkhP motif at lower pHi in cells and that pH-regulated binding and activity are dependent on a conserved histidine (His122) in the DBD. RNA-seq with FOXC2 also reveals pH-dependent differences in enriched promoter motifs. Our findings identify pH-regulated transcription factor-DNA binding selectivity with relevance to how pHi dynamics can regulate gene expression for myriad cell behaviours.
Collapse
|
5
|
Rysenkova KD, Gaboriaud J, Fokin AI, Toubiana R, Bense A, Mirdass C, Jin M, Ho MCN, Glading E, Vacher S, Courtois L, Bièche I, Gautreau AM. PI 3-Kinase and the Histone Methyl-Transferase KMT2D Collaborate to Induce Arp2/3-Dependent Migration of Mammary Epithelial Cells. Cells 2024; 13:876. [PMID: 38786098 PMCID: PMC11119607 DOI: 10.3390/cells13100876] [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: 02/13/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer develops upon sequential acquisition of driver mutations in mammary epithelial cells; however, how these mutations collaborate to transform normal cells remains unclear in most cases. We aimed to reconstitute this process in a particular case. To this end, we combined the activated form of the PI 3-kinase harboring the H1047R mutation with the inactivation of the histone lysine methyl-transferase KMT2D in the non-tumorigenic human mammary epithelial cell line MCF10A. We found that PI 3-kinase activation promoted cell-cycle progression, especially when growth signals were limiting, as well as cell migration, both in a collective monolayer and as single cells. Furthermore, we showed that KMT2D inactivation had relatively little influence on these processes, except for single-cell migration, which KMT2D inactivation promoted in synergy with PI 3-kinase activation. The combination of these two genetic alterations induced expression of the ARPC5L gene that encodes a subunit of the Arp2/3 complex. ARPC5L depletion fully abolished the enhanced migration persistence exhibited by double-mutant cells. Our reconstitution approach in MCF10A has thus revealed both the cell function and the single-cell migration, and the underlying Arp2/3-dependent mechanism, which are synergistically regulated when KMT2D inactivation is combined with the activation of the PI 3-kinase.
Collapse
Affiliation(s)
- Karina D. Rysenkova
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Julia Gaboriaud
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Artem I. Fokin
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Raphaëlle Toubiana
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Alexandre Bense
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Camil Mirdass
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Mélissa Jin
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Minh Chau N. Ho
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Elizabeth Glading
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| | - Sophie Vacher
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Laura Courtois
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Ivan Bièche
- Pharmacogenomics Unit, Department of Genetics, Institut Curie, Paris Descartes University, 75005 Paris, France; (S.V.); (L.C.); (I.B.)
| | - Alexis M. Gautreau
- Laboratoire de Biologie Structurale de la Cellule, CNRS UMR7654, Ecole Polytechnique, Institut Polytechnique de Paris, 91128 Palaiseau Cedex, France; (K.D.R.); (J.G.); (A.I.F.); (R.T.); (A.B.); (C.M.); (M.J.); (M.C.N.H.); (E.G.)
| |
Collapse
|
6
|
Jamali M, Barar E, Shi J. Unveiling the Molecular Landscape of Pancreatic Ductal Adenocarcinoma: Insights into the Role of the COMPASS-like Complex. Int J Mol Sci 2024; 25:5069. [PMID: 38791111 PMCID: PMC11121229 DOI: 10.3390/ijms25105069] [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: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is poised to become the second leading cause of cancer-related death by 2030, necessitating innovative therapeutic strategies. Genetic and epigenetic alterations, including those involving the COMPASS-like complex genes, have emerged as critical drivers of PDAC progression. This review explores the genetic and epigenetic landscape of PDAC, focusing on the role of the COMPASS-like complex in regulating chromatin accessibility and gene expression. Specifically, we delve into the functions of key components such as KDM6A, KMT2D, KMT2C, KMT2A, and KMT2B, highlighting their significance as potential therapeutic targets. Furthermore, we discuss the implications of these findings for developing novel treatment modalities for PDAC.
Collapse
Affiliation(s)
- Marzieh Jamali
- Department of Pathology & Clinical Labs, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erfaneh Barar
- Liver and Pancreatobiliary Diseases Research Center, Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Jiaqi Shi
- Department of Pathology & Clinical Labs, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Xu D, Bewicke-Copley F, Close K, Okosun J, Gale RP, Apperley J, Weinstock DM, Wendel HG, Fitzgibbon J. Targeting lysine demethylase 5 (KDM5) in mantle cell lymphoma. Blood Cancer J 2024; 14:29. [PMID: 38351059 PMCID: PMC10864367 DOI: 10.1038/s41408-024-00999-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024] Open
Affiliation(s)
- Danmei Xu
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK.
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK.
- Centre for Haematology, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK.
- Oxford Cancer and Haematology centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK.
| | - Findlay Bewicke-Copley
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| | - Karina Close
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| | - Robert Peter Gale
- Centre for Haematology, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Jane Apperley
- Centre for Haematology, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Merck and Co., Rahway, NJ, USA
| | - Hans-Guido Wendel
- Memorial Sloan-Kettering Cancer Center, Cancer Biology & Genetics, New York, NY, 10065, USA
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Sq, London, UK
| |
Collapse
|
8
|
Golden CS, Williams S, Serrano MA. Molecular insights of KMT2D and clinical aspects of Kabuki syndrome type 1. Birth Defects Res 2023; 115:1809-1824. [PMID: 37158694 PMCID: PMC10845236 DOI: 10.1002/bdr2.2183] [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: 01/25/2023] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Kabuki syndrome type 1 (KS1), a rare multisystem congenital disorder, presents with characteristic facial features, intellectual disability, persistent fetal fingertip pads, skeletal abnormalities, and postnatal growth delays. KS1 results from pathogenic variants in the KMT2D gene, which encodes a histone methyltransferase protein involved in chromatin remodeling, promoter and enhancer regulation, and scaffold formation during early development. KMT2D also mediates cell signaling pathways, responding to external stimuli and organizing effector protein assembly. Research on KMT2D's molecular mechanisms in KS1 has primarily focused on its histone methyltransferase activity, leaving a gap in understanding the methyltransferase-independent roles in KS1 clinical manifestations. METHODS This scoping review examines KMT2D's role in gene expression regulation across various species, cell types, and contexts. We analyzed human pathogenic KMT2D variants using publicly available databases and compared them to research organism models of KS1. We also conducted a systematic search of healthcare and governmental databases for clinical trials, studies, and therapeutic approaches. RESULTS Our review highlights KMT2D's critical roles beyond methyltransferase activity in diverse cellular contexts and conditions. We identified six distinct groups of KMT2D as a cell signaling mediator, including evidence of methyltransferase-dependent and -independent activity. A comprehensive search of the literature, clinical databases, and public registries emphasizes the need for basic research on KMT2D's functional complexity and longitudinal studies of KS1 patients to establish objective outcome measurements for therapeutic development. CONCLUSION We discuss how KMT2D's role in translating external cellular communication can partly explain the clinical heterogeneity observed in KS1 patients. Additionally, we summarize the current molecular diagnostic approaches and clinical trials targeting KS1. This review is a resource for patient advocacy groups, researchers, and physicians to support KS1 diagnosis and therapeutic development.
Collapse
Affiliation(s)
- Carly S Golden
- Center for Regenerative Medicine, Section of Vascular Biology, Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Saylor Williams
- Center for Regenerative Medicine, Section of Vascular Biology, Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Maria A Serrano
- Center for Regenerative Medicine, Section of Vascular Biology, Department of Medicine, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Kashiwabara L, Pirard L, Debier C, Crocker D, Khudyakov J. Effects of cortisol, epinephrine, and bisphenol contaminants on the transcriptional landscape of marine mammal blubber. Am J Physiol Regul Integr Comp Physiol 2023; 325:R504-R522. [PMID: 37602383 DOI: 10.1152/ajpregu.00165.2023] [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: 07/02/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
Top ocean predators such as marine mammals are threatened by intensifying anthropogenic activity, and understanding the combined effects of multiple stressors on their physiology is critical for conservation efforts. We investigated potential interactions between stress hormones and bisphenol contaminants in a model marine mammal, the northern elephant seal (NES). We exposed precision-cut adipose tissue slices (PCATS) from blubber of weaned NES pups to cortisol (CORT), epinephrine (EPI), bisphenol A (BPA), bisphenol S (BPS), or their combinations (CORT-EPI, BPA-EPI, and BPS-EPI) ex vivo and identified hundreds of genes that were differentially regulated in response to these treatments. CORT altered expression of genes associated with lipolysis and adipogenesis, whereas EPI and CORT-EPI-regulated genes were associated with responses to hormones, lipid and protein turnover, immune function, and transcriptional and epigenetic regulation of gene expression, suggesting that EPI has wide-ranging and prolonged impacts on the transcriptional landscape and function of blubber. Bisphenol treatments alone had a weak impact on gene expression compared with stress hormones. However, the combination of EPI with bisphenols altered expression of genes associated with inflammation, cell stress, DNA damage, regulation of nuclear hormone receptor activity, cell cycle, mitochondrial function, primary ciliogenesis, and lipid metabolism in blubber. Our results suggest that CORT, EPI, bisphenols, and their combinations impact cellular, immune, and metabolic homeostasis in marine mammal blubber, which may affect the ability of marine mammals to sustain prolonged fasting during reproduction and migration, renew tissues, and mount appropriate responses to immune challenges and additional stressors.
Collapse
Affiliation(s)
- Lauren Kashiwabara
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States
| | - Laura Pirard
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - Cathy Debier
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la Neuve, Belgium
| | - Daniel Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California, United States
| | - Jane Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States
| |
Collapse
|
10
|
Shayeghpour A, Forghani-Ramandi MM, Solouki S, Hosseini A, Hosseini P, Khodayar S, Hasani M, Aghajanian S, Siami Z, Zarei Ghobadi M, Mozhgani SH. Identification of novel miRNAs potentially involved in the pathogenesis of adult T-cell leukemia/lymphoma using WGCNA followed by RT-qPCR test of hub genes. Infect Agent Cancer 2023; 18:12. [PMID: 36841815 PMCID: PMC9968414 DOI: 10.1186/s13027-023-00492-0] [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: 08/11/2022] [Accepted: 02/17/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Adult T-cell Lymphoma/Leukemia (ATLL) is characterized by the malignant proliferation of T-cells in Human T-Lymphotropic Virus Type 1 and a high mortality rate. Considering the emerging roles of microRNAs (miRNAs) in various malignancies, the analysis of high-throughput miRNA data employing computational algorithms helps to identify potential biomarkers. METHODS Weighted gene co-expression network analysis was utilized to analyze miRNA microarray data from ATLL and healthy uninfected samples. To identify miRNAs involved in the progression of ATLL, module preservation analysis was used. Subsequently, based on the target genes of the identified miRNAs, the STRING database was employed to construct protein-protein interaction networks (PPIN). Real-time quantitative PCR was also performed to validate the expression of identified hub genes in the PPIN network. RESULTS After constructing co-expression modules and then performing module preservation analysis, four out of 15 modules were determined as ATLL-specific modules. Next, the hub miRNA including hsa-miR-18a-3p, has-miR-187-5p, hsa-miR-196a-3p, and hsa-miR-346 were found as hub miRNAs. The protein-protein interaction networks were constructed for the target genes of each hub miRNA and hub genes were identified. Among them, UBB, RPS15A, and KMT2D were validated by Reverse-transcriptase PCR in ATLL patients. CONCLUSION The results of the network analysis of miRNAs and their target genes revealed the major players in the pathogenesis of ATLL. Further studies are required to confirm the role of these molecular factors and to discover their potential benefits as treatment targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Ali Shayeghpour
- grid.411705.60000 0001 0166 0922School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Setayesh Solouki
- grid.411705.60000 0001 0166 0922School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Amin Hosseini
- Department of Computer, Faculty of Engineering, Raja University, Qazvin, Iran
| | - Parastoo Hosseini
- grid.411705.60000 0001 0166 0922Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran ,grid.411705.60000 0001 0166 0922Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Khodayar
- grid.411705.60000 0001 0166 0922Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahsa Hasani
- grid.411705.60000 0001 0166 0922School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Sepehr Aghajanian
- grid.411705.60000 0001 0166 0922School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Zeinab Siami
- grid.411705.60000 0001 0166 0922Department of Infectious Diseases, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran. .,Non-Communicable Disease Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
11
|
Pan Y, Han H, Hu H, Wang H, Song Y, Hao Y, Tong X, Patel AS, Misirlioglu S, Tang S, Huang HY, Geng K, Chen T, Karatza A, Sherman F, Labbe KE, Yang F, Chafitz A, Peng C, Guo C, Moreira AL, Velcheti V, Lau SCM, Sui P, Chen H, Diehl JA, Rustgi AK, Bass AJ, Poirier JT, Zhang X, Ji H, Zhang H, Wong KK. KMT2D deficiency drives lung squamous cell carcinoma and hypersensitivity to RTK-RAS inhibition. Cancer Cell 2023; 41:88-105.e8. [PMID: 36525973 PMCID: PMC10388706 DOI: 10.1016/j.ccell.2022.11.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/06/2022] [Accepted: 11/22/2022] [Indexed: 12/16/2022]
Abstract
Lung squamous cell carcinoma (LUSC) represents a major subtype of lung cancer with limited treatment options. KMT2D is one of the most frequently mutated genes in LUSC (>20%), and yet its role in LUSC oncogenesis remains unknown. Here, we identify KMT2D as a key regulator of LUSC tumorigenesis wherein Kmt2d deletion transforms lung basal cell organoids to LUSC. Kmt2d loss increases activation of receptor tyrosine kinases (RTKs), EGFR and ERBB2, partly through reprogramming the chromatin landscape to repress the expression of protein tyrosine phosphatases. These events provoke a robust elevation in the oncogenic RTK-RAS signaling. Combining SHP2 inhibitor SHP099 and pan-ERBB inhibitor afatinib inhibits lung tumor growth in Kmt2d-deficient LUSC murine models and in patient-derived xenografts (PDXs) harboring KMT2D mutations. Our study identifies KMT2D as a pivotal epigenetic modulator for LUSC oncogenesis and suggests that KMT2D loss renders LUSC therapeutically vulnerable to RTK-RAS inhibition.
Collapse
Affiliation(s)
- Yuanwang Pan
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Han Han
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Hua Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yueqiang Song
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Hao
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA; Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, NY, USA
| | - Xinyuan Tong
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ayushi S Patel
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Selim Misirlioglu
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Sittinon Tang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Hsin-Yi Huang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Ke Geng
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Angeliki Karatza
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Fiona Sherman
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Kristen E Labbe
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Fan Yang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Alison Chafitz
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Chengwei Peng
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Chenchen Guo
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Andre L Moreira
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Sally C M Lau
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - J Alan Diehl
- Department of Biochemistry, Case Western Reserve University and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Adam J Bass
- Herbert Irving Comprehensive Cancer Center, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - John T Poirier
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Xiaoyang Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Hua Zhang
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA; Hillman Cancer Center, UPMC, Pittsburgh, PA 15232, USA; Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
12
|
Pérez-Montiel MD, Cerrato-Izaguirre D, Sánchez-Pérez Y, Diaz-Chavez J, Cortés-González CC, Rubio JA, Jiménez-Ríos MA, Herrera LA, Scavuzzo A, Meneses-García A, Hernández-Martínez R, Vaca-Paniagua F, Ramírez A, Orozco A, Cantú-de-León D, Prada D. Mutational Landscape of Bladder Cancer in Mexican Patients: KMT2D Mutations and chr11q15.5 Amplifications Are Associated with Muscle Invasion. Int J Mol Sci 2023; 24:ijms24021092. [PMID: 36674608 PMCID: PMC9866210 DOI: 10.3390/ijms24021092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/27/2022] [Accepted: 12/03/2022] [Indexed: 01/09/2023] Open
Abstract
Bladder cancer (BC) is the most common neoplasm of the urinary tract, which originates in the epithelium that covers the inner surface of the bladder. The molecular BC profile has led to the development of different classifications of non-muscle invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC). However, the genomic BC landscape profile of the Mexican population, including NMIBC and MIBC, is unknown. In this study, we aimed to identify somatic single nucleotide variants (SNVs) and copy number variations (CNVs) in Mexican patients with BC and their associations with clinical and pathological characteristics. We retrospectively evaluated 37 patients treated between 2012 and 2021 at the National Cancer Institute-Mexico (INCan). DNA samples were obtained from paraffin-embedded tumor tissues and exome sequenced. Strelka2 and Lancet packages were used to identify SNVs and insertions or deletions. FACETS was used to determine CNVs. We found a high frequency of mutations in TP53 and KMT2D, gains in 11q15.5 and 19p13.11-q12, and losses in 7q11.23. STAG2 mutations and 1q11.23 deletions were also associated with NMIBC and low histologic grade.
Collapse
Affiliation(s)
- María D. Pérez-Montiel
- Departamento de Patología, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Dennis Cerrato-Izaguirre
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N. (CINVESTAV), Avenida Instituto Politécnico Nacional No. 2508, Mexico City 07360, Mexico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Jose Diaz-Chavez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Carlo César Cortés-González
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Jairo A. Rubio
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Miguel A. Jiménez-Ríos
- Departamento de Urología, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Luis A. Herrera
- Dirección General, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico
| | - Anna Scavuzzo
- Departamento de Urología, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | | | - Ricardo Hernández-Martínez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Felipe Vaca-Paniagua
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Tlalnepantla 54090, Mexico
| | - Andrea Ramírez
- Unidad de Apoyo Molecular a la Investigación Clínica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - Alicia Orozco
- Unidad de Apoyo Molecular a la Investigación Clínica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
| | - David Cantú-de-León
- Dirección de Investigación, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Correspondence: (D.C.-d.-L.); (D.P.); Tel.: +52-553693-5200 (ext. 241) (D.P.)
| | - Diddier Prada
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Mexico City 14080, Mexico
- Department of Environmental Health Sciences, Mailman School of Public Health, New York, NY 10032, USA
- Correspondence: (D.C.-d.-L.); (D.P.); Tel.: +52-553693-5200 (ext. 241) (D.P.)
| |
Collapse
|
13
|
Aukema SM, Glaser S, van den Hout MFCM, Dahlum S, Blok MJ, Hillmer M, Kolarova J, Sciot R, Schott DA, Siebert R, Stumpel CTRM. Molecular characterization of an embryonal rhabdomyosarcoma occurring in a patient with Kabuki syndrome: report and literature review in the light of tumor predisposition syndromes. Fam Cancer 2023; 22:103-118. [PMID: 35856126 PMCID: PMC9829644 DOI: 10.1007/s10689-022-00306-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 01/13/2023]
Abstract
Kabuki syndrome is a well-recognized syndrome characterized by facial dysmorphism and developmental delay/intellectual disability and in the majority of patients a germline variant in KMT2D is found. As somatic KMT2D variants can be found in 5-10% of tumors a tumor predisposition in Kabuki syndrome is discussed. So far less than 20 patients with Kabuki syndrome and a concomitant malignancy have been published. Here we report on a female patient with Kabuki syndrome and a c.2558_2559delCT germline variant in KMT2D who developed an embryonal rhabdomyosarcoma (ERMS) at 10 years. On tumor tissue we performed DNA-methylation profiling and exome sequencing (ES). Copy number analyses revealed aneuploidies typical for ERMS including (partial) gains of chromosomes 2, 3, 7, 8, 12, 15, and 20 and 3 focal deletions of chromosome 11p. DNA methylation profiling mapped the case to ERMS by a DNA methylation-based sarcoma classifier. Sequencing suggested gain of the wild-type KMT2D allele in the trisomy 12. Including our patient literature review identified 18 patients with Kabuki syndrome and a malignancy. Overall, the landscape of malignancies in patients with Kabuki syndrome was reminiscent of that of the pediatric population in general. Histopathological and molecular data were only infrequently reported and no report included next generation sequencing and/or DNA-methylation profiling. Although we found no strong arguments pointing towards KS as a tumor predisposition syndrome, based on the small numbers any relation cannot be fully excluded. Further planned studies including profiling of additional tumors and long term follow-up of KS-patients into adulthood could provide further insights.
Collapse
Affiliation(s)
- Sietse M Aukema
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - Selina Glaser
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Mari F C M van den Hout
- Department of Pathology, Research Institute GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sonja Dahlum
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Marinus J Blok
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Morten Hillmer
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Julia Kolarova
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Raf Sciot
- Department of Pathology, University Hospital, University of Leuven, 3000, Louvain, Belgium
| | - Dina A Schott
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
- Department of Pediatrics, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Constance T R M Stumpel
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
- Department of Clinical Genetics and GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
14
|
Congur I, Koni E, Onat OE, Tokcaer Keskin Z. Meta-analysis of commonly mutated genes in leptomeningeal carcinomatosis. PeerJ 2023; 11:e15250. [PMID: 37096065 PMCID: PMC10122459 DOI: 10.7717/peerj.15250] [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: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 04/26/2023] Open
Abstract
Background Leptomeningeal carcinomatosis (LMC) is a rare type of cancer that settles at the meninges through metastasis of non-small cell lung cancer (NSCLC), breast cancer and melanoma. The molecular mechanism underlying LMC is not known, therefore molecular studies investigating the development of LMC are needed. Here, we aimed to identify commonly mutated genes in LMC caused by NSCLC, breast cancer, and melanoma using an in-slico approach and their interactions using integrated bioinformatic approaches/tools in this meta-analysis. Methods We conducted a meta-analysis using information from 16 studies that included different sequencing techniques of patients with LMC caused by three different primary cancers: breast cancer, NSCLC, and melanoma. All studies that assessed mutation information from patients with LMC were searched in PubMed, from their inception to February, 16 2022. Studies that performed NGS on LMC patients with NSCLC, breast cancer, or melanoma were included, while studies that did not apply NGS to CSF samples, did not provide information on altered genes, were reviews, editorials, or conference abstracts, or whose main goal was the detection of malignancies were all excluded. We identified commonly mutated genes in all three types of cancer. Next, we constructed a protein-protein interaction network, then performed pathway enrichment analysis. We searched National Institutes of Health (NIH) and Drug-Gene Interaction Database (DGIdb) to find candidate drugs. Results We found that TP53, PTEN, PIK3CA, IL7R, and KMT2D genes were commonly mutated genes in all three types of cancer via our meta-analysis that consisted out of 16 studies. Our pathway enrichment analysis showed that all five genes were primarily associated with regulation of cell communication and signaling, and cell proliferation. Other enriched pathways included regulation of apoptotic processes of leukocytes and fibroblasts, macroautophagy and growth. According to our drug search we found candidate drugs; Everolimus, Bevacizumab and Temozolomide, which interact with these five genes. Conclusion In conclusion, a total of 96 mutated genes in LMC were investigated via meta-analysis. Our findings suggested vital roles of TP53, PTEN, PIK3CA, KMT2D, and IL7R, which can provide insight into the molecular basis of LMC development and paving the door to the development of new targeted medicine and will encourage molecular biologists to seek biological evidence.
Collapse
Affiliation(s)
- Irem Congur
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Ekin Koni
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Onur Emre Onat
- Department of Genome Studies, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
- Department of Molecular Biology, Institute of Life Sciences and Biotechnology, Bezmialem Foundation University, Istanbul, Turkey
| | - Zeynep Tokcaer Keskin
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics Faculty of Engineering and Natural Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| |
Collapse
|
15
|
Lau SCM, Pan Y, Velcheti V, Wong KK. Squamous cell lung cancer: Current landscape and future therapeutic options. Cancer Cell 2022; 40:1279-1293. [PMID: 36270277 DOI: 10.1016/j.ccell.2022.09.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/19/2022] [Accepted: 09/27/2022] [Indexed: 01/09/2023]
Abstract
Squamous cell lung cancers (lung squamous cell carcinomas [LUSCs]) are associated with high mortality and a lack of therapies specific to this disease. Although recurrent molecular aberrations are present in LUSCs, efforts to develop targeted therapies against receptor tyrosine kinases, signaling transduction, and cell cycle checkpoints in LUSCs were met with significant challenges. The present therapeutic landscape focuses on epigenetic therapies to modulate the expression of lineage-dependent survival pathways and undruggable oncogenes. Another important therapeutic approach is to exploit metabolic vulnerabilities unique to LUSCs. These novel therapies may synergize with immune checkpoint inhibitors in the right therapeutic context. For example, the recognition that alterations in KEAP1-NFE2L2 in LUSCs affected antitumor immune responses created unique opportunities for targeted, metabolic, and immune combinations. This article provides a perspective on how lessons learned from the past influence the current therapeutic landscape and opportunities for future drug development for LUSCs.
Collapse
Affiliation(s)
- Sally C M Lau
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Yuanwang Pan
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Vamsidhar Velcheti
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA
| | - Kwok Kin Wong
- Department of Medical Oncology, Laura & Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, Smilow Building 10th Floor, Suite 1001, New York, NY 10016, USA.
| |
Collapse
|
16
|
Epigenetic factor competition reshapes the EMT landscape. Proc Natl Acad Sci U S A 2022; 119:e2210844119. [PMID: 36215492 PMCID: PMC9586264 DOI: 10.1073/pnas.2210844119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The emergence of and transitions between distinct phenotypes in isogenic cells can be attributed to the intricate interplay of epigenetic marks, external signals, and gene-regulatory elements. These elements include chromatin remodelers, histone modifiers, transcription factors, and regulatory RNAs. Mathematical models known as gene-regulatory networks (GRNs) are an increasingly important tool to unravel the workings of such complex networks. In such models, epigenetic factors are usually proposed to act on the chromatin regions directly involved in the expression of relevant genes. However, it has been well-established that these factors operate globally and compete with each other for targets genome-wide. Therefore, a perturbation of the activity of a regulator can redistribute epigenetic marks across the genome and modulate the levels of competing regulators. In this paper, we propose a conceptual and mathematical modeling framework that incorporates both local and global competition effects between antagonistic epigenetic regulators, in addition to local transcription factors, and show the counterintuitive consequences of such interactions. We apply our approach to recent experimental findings on the epithelial-mesenchymal transition (EMT). We show that it can explain the puzzling experimental data, as well as provide verifiable predictions.
Collapse
|
17
|
Wang L, Tang J, Feng J, Huang Y, Cheng Y, Xu H, Miao Y. Case report: A rare case of coexisting Waldenstrom Macroglobulinemia and B-cell acute lymphoblastic leukemia with KMT2D and MECOM mutations. Front Immunol 2022; 13:1001482. [PMID: 36325357 PMCID: PMC9618799 DOI: 10.3389/fimmu.2022.1001482] [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: 07/23/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background Waldenstrom Macroglobulinemia (WM) is a rare and indolent lymphoma of B-cell origin characterized by elevated monoclonal IgM, with MYD88L265P mutation and CXCR4 mutation as common molecular alterations. B-cell Acute Lymphoblastic Leukemia (B-ALL) is clinically heterogeneous, characterized by abnormal proliferation and aggregation of immature lymphocytes in the bone marrow and lymphoid tissue. WM and ALL are hematologic malignancies of B-cell origin with completely different clinical manifestations and biological features. KMT2D and MECOM mutations are very rare in ALL and usually indicate poor disease prognosis. The coexistence of WM and ALL with KMT2D and MECOM mutations have not been reported. Case presentation A 74-year-old female patient was diagnosed with WM in July 2018 and received four cycles of chemotherapy of bortezomib and dexamethasone. In November 2018, she received immunomodulator thalidomide as maintenance therapy. In November 2020, Bruton’s Tyrosine Kinase inhibitors (BTKi) has been introduced into the Chinese market and she took zanubrutinib orally at a dose of 80 mg per day. The disease remained in remission. In December 2021, she presented with multiple enlarged lymph nodes throughout the body. Bone marrow and next-generation sequencing (NGS) suggested the coexistence of WM and B-ALL with KMT2D and MECOM mutations. The patient was treated with zanubrutinib in combination with vincristine and dexamethasone, after which she developed severe myelosuppression and septicemia. The patient finally got remission. Due to the patient’s age and poor status, she refused intravenous chemotherapy and is currently treated with zanubrutinib. Conclusions The coexistence of WM and B-ALL is very rare and has not been reported. The presence of both KMT2D and MECOM mutations predicts a poor prognosis and the possibility of insensitivity to conventional treatment options. BTKi achieves its anti-tumor effects by inhibiting BTK activation and blocking a series of malignant transformations in B-cell tumors. In addition, it also acts on T-cell immunity and tumor microenvironment. Combination therapy based on BTKi may improve the prognosis of this patient.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Jiao Tang
- Department of Neurology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Jun Feng
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Yongfen Huang
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Yuexin Cheng
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Hao Xu
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
| | - Yuqing Miao
- Department of Hematology, The First People’s Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, China
- *Correspondence: Yuqing Miao,
| |
Collapse
|
18
|
Barry KK, Tsaparlis M, Hoffman D, Hartman D, Adam MP, Hung C, Bodamer OA. From Genotype to Phenotype-A Review of Kabuki Syndrome. Genes (Basel) 2022; 13:1761. [PMID: 36292647 PMCID: PMC9601850 DOI: 10.3390/genes13101761] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022] Open
Abstract
Kabuki syndrome (KS) is a rare neuro-developmental disorder caused by variants in genes of histone modification, including KMT2D and KDM6A. This review assesses our current understanding of KS, which was originally named Niikawa-Kuroki syndrome, and aims to guide surveillance and medical care of affected individuals as well as identify gaps in knowledge and unmet patient needs. Ovid MEDLINE and EMBASE databases were searched from 1981 to 2021 to identify reports related to genotype and systems-based phenotype characterization of KS. A total of 2418 articles were retrieved, and 152 were included in this review, representing a total of 1369 individuals with KS. Genotype, phenotype, and the developmental and behavioral profile of KS are reviewed. There is a continuous clinical phenotype spectrum associated with KS with notable variability between affected individuals and an emerging genotype-phenotype correlation. The observed clinical variability may be attributable to differences in genotypes and/or unknown genetic and epigenetic factors. Clinical management is symptom oriented, fragmented, and lacks established clinical care standards. Additional research should focus on enhancing understanding of the burden of illness, the impact on quality of life, the adult phenotype, life expectancy and development of standard-of-care guidelines.
Collapse
Affiliation(s)
- Kelly K. Barry
- Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | - Margaret P. Adam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Christina Hung
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Olaf A. Bodamer
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|
19
|
Levescot A, Malamut G, Cerf-Bensussan N. Immunopathogenesis and environmental triggers in coeliac disease. Gut 2022; 71:gutjnl-2021-326257. [PMID: 35879049 PMCID: PMC9554150 DOI: 10.1136/gutjnl-2021-326257] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/07/2022] [Indexed: 12/21/2022]
Abstract
Coeliac disease (CD) is a frequent immune enteropathy induced by gluten in genetically predisposed individuals. Its pathogenesis has been extensively studied and CD has emerged as a model disease to decipher how the interplay between environmental and genetic factors can predispose to autoimmunity and promote lymphomagenesis. The keystone event is the activation of a gluten-specific immune response that is driven by molecular interactions between gluten, the indispensable environmental factor, HLA-DQ2/8, the main predisposing genetic factor and transglutaminase 2, the CD-specific autoantigen. The antigluten response is however not sufficient to induce epithelial damage which requires the activation of cytotoxic CD8+ intraepithelial lymphocytes (IEL). In a plausible scenario, cooperation between cytokines released by gluten-specific CD4+ T cells and interleukin-15 produced in excess in the coeliac gut, licenses the autoimmune-like attack of the gut epithelium, likely via sustained activation of the Janus kinase-signal transducer and activator of transcription (JAK/STAT) pathway in IEL. Demonstration that lymphomas complicating CD arise from IEL that have acquired gain-of-function JAK1 or STAT3 mutations stresses the key role of this pathway and explains how gluten-driven chronic inflammation may promote this rare but most severe complication. If our understanding of CD pathogenesis has considerably progressed, several questions and challenges remain. One unsolved question concerns the considerable variability in disease penetrance, severity and presentation, pointing to the role of additional genetic and environmental factors that remain however uneasy to untangle and hierarchize. A current challenge is to transfer the considerable mechanistic insight gained into CD pathogenesis into benefits for the patients, notably to alleviate the gluten-free diet, a burden for many patients.
Collapse
Affiliation(s)
- Anais Levescot
- Université Paris Cité, Institut Imagine, INSERM UMR1163, Laboratory Intestinal Immunity, Paris, France
| | - Georgia Malamut
- Université Paris Cité, Institut Imagine, INSERM UMR1163, Laboratory Intestinal Immunity, Paris, France
- Université Paris Cité, APHP Centre, Gastroenterology Department, Hôpital Cochin, Paris, France
| | - Nadine Cerf-Bensussan
- Université Paris Cité, Institut Imagine, INSERM UMR1163, Laboratory Intestinal Immunity, Paris, France
| |
Collapse
|
20
|
Wang X, Li R, Wu L, Chen Y, Liu S, Zhao H, Wang Y, Wang L, Shao Z. Histone methyltransferase KMT2D cooperates with MEF2A to promote the stem-like properties of oral squamous cell carcinoma. Cell Biosci 2022; 12:49. [PMID: 35477537 PMCID: PMC9044881 DOI: 10.1186/s13578-022-00785-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 04/11/2022] [Indexed: 12/03/2022] Open
Abstract
Background Epigenetic reprogramming is involved in multiple steps of human cancer evolution and is mediated by a variety of chromatin-modifying enzymes. Specifically, the histone lysine methyltransferase KMT2D is among the most frequently mutated genes in oral squamous cell carcinoma (OSCC). However, the mechanisms by which KMT2D affects the development of OSCC remain unclear. Results In the present study, we found that the expression of KMT2D was elevated in OSCC compared to paracancerous specimens and was correlated with a more advanced tumor grade. More importantly, knockdown of KMT2D impaired their reconstitution in patient-derived organoids and decreased the expression of CD133 and β-catenin in OSCC cells. In in vitro and in vivo models, knockdown of KMT2D reduced the colony formation, migration and invasion abilities of OSCC cells and delayed tumor growth. Mechanistically, the dual-luciferase reporter and co-immunoprecipitation assays in two individual OSCC cell lines indicated that KMT2D may cooperate with MEF2A to promote the transcription activity of CTNNB1, thereby enhancing WNT signaling. Conclusion The upregulation of KMT2D contributes to stem-like properties in OSCC cells by sustaining the MEF2A-mediated transcriptional activity of CTNNB1. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00785-8.
Collapse
Affiliation(s)
- Xinmiao Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Rui Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Luping Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Yang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Shaopeng Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Hui Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Yifan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China
| | - Lin Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Zhe Shao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430089, China. .,Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Yang MZ, Li L, Wei H, Liu BC, Liu KQ, Li DP, Zhang L, Yang RC, Mi YC, Wang JX, Wang Y. [Clinical and genetic characteristics of patients with newly diagnosed acute promyelocytic leukemia: a single-center retrospective of 790 cases]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:336-341. [PMID: 35680634 PMCID: PMC9189486 DOI: 10.3760/cma.j.issn.0253-2727.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Indexed: 11/05/2022]
Abstract
Objective: To retrospectively analyze the data of Chinese patients with newly diagnosed acute promyelocytic leukemia (APL) to preliminarily discuss the clinical and cytogenetic characteristics. Methods: From February 2004 to June 2020, patients with newly diagnosed APL aged ≥ 15 years who were admitted to the Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College were chosen. Clinical and laboratory features were retrospectively analyzed. Results: A total of 790 cases were included, with a male to female ratio of 1.22. The median age of the patients was 41 (15-76) years. Patients aged between 20 and 59 predominated, with 632 patients (80%) of 790 patients classified as low and intermediate risk and 158 patients (20%) of 790 patients classified as high risk. The white blood cell, platelet, and hemoglobin levels at diagnosis were 2.3 (0.1-176.1) ×10(9)/L, 29.5 (2.0-1220.8) ×10(9)/L, and 89 (15-169) g/L, respectively, and 4.8% of patients were complicated with psoriasis. The long-form type of PML-RARα was most commonly seen in APL, accounting for 58%. Both APTT extension (10.3%) and creatinine>14 mg/L (1%) are rarely seen in patients at diagnosis. Cytogenetics was performed in 715 patients with newly diagnosed APL. t (15;17) with additional chromosomal abnormalities were found in 155 patients, accounting for 21.7%; among which, +8 was most frequently seen. A complex karyotype was found in 64 (9.0%) patients. Next-generation sequencing was performed in 178 patients, and 113 mutated genes were discovered; 75 genes had an incidence rate>1%. FLT3 was the most frequently seen, which accounted for 44.9%, and 20.8% of the 178 patients present with FLT3-ITD. Conclusions: Patients aged 20-59 years are the most common group with newly diagnosed APL. No obvious difference was found in the ratio of males to females. In terms of risk stratification, patients divided into low and intermediate risk predominate. t (15;17) with additional chromosomal abnormalities accounted for 21% of 715 patients, in which +8 was most commonly seen. The long-form subtype was most frequently seen in PML-RARα-positive patients, and FLT3 was most commonly seen in the mutation spectrum of APL.
Collapse
Affiliation(s)
- M Z Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - L Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - H Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - B C Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - K Q Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - D P Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - L Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - R C Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y C Mi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - J X Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
22
|
Niu Y, Wang G, Li Y, Guo W, Guo Y, Dong Z. LncRNA FOXP4-AS1 Promotes the Progression of Esophageal Squamous Cell Carcinoma by Interacting With MLL2/H3K4me3 to Upregulate FOXP4. Front Oncol 2022; 11:773864. [PMID: 34970490 PMCID: PMC8712759 DOI: 10.3389/fonc.2021.773864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/19/2021] [Indexed: 01/16/2023] Open
Abstract
Malignant tumors are a grave threat to human health. Esophageal squamous cell carcinoma (ESCC) is a common gastrointestinal malignant tumor. China has a high incidence of ESCC, and its morbidity and mortality are higher than the global average. Increasingly, studies have shown that long noncoding RNAs (lncRNAs) play a vital function in the occurrence and development of tumors. Although the biological function of FOXP4-AS1 has been demonstrated in various tumors, the potential molecular mechanism of FOXP4-AS1 in ESCC is still poorly understood. The expression of FOXP4 and FOXP4-AS1 was detected in ESCC by quantitative real-time PCR (qRT–PCR) or SP immunohistochemistry (IHC). shRNA was used to silence gene expression. Apoptosis, cell cycle, MTS, colony formation, invasion and migration assays were employed to explore the biological functions of FOXP4 and FOXP4-AS1. The potential molecular mechanism of FOXP4-AS1 in ESCC was determined by dual-luciferase reporter, RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP). Here, we demonstrated that FOXP4-AS1 was significantly increased in ESCC tissues and cell lines, associated with lymph node metastasis and TNM staging. Cell function experiments showed that FOXP4-AS1 promoted the proliferation, invasion and migration ability of ESCC cells. The expression of FOXP4-AS1 and FOXP4 in ESCC tissues was positively correlated. Further research found that FOXP4-AS1, upregulated in ESCC, promotes FOXP4 expression by enriching MLL2 and H3K4me3 in the FOXP4 promoter through a “molecular scaffold”. Moreover, FOXP4, a transcription factor of β-catenin, promotes the transcription of β-catenin and ultimately leads to the malignant progression of ESCC. Finally, FOXP4-AS1 may be a new therapeutic target for ESCC.
Collapse
Affiliation(s)
- Yunfeng Niu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gaoyan Wang
- Experimental Center, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yan Li
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanli Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiming Dong
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
23
|
Laukhtina E, Lemberger U, Bruchbacher A, Ilijazi D, Korn S, Berndl F, D’Andrea D, Susani M, Enikeev D, Compérat E, Shariat SF, Hassler MR. Expression Analysis and Mutational Status of Histone Methyltransferase KMT2D at Different Upper Tract Urothelial Carcinoma Locations. J Pers Med 2021; 11:1147. [PMID: 34834500 PMCID: PMC8625702 DOI: 10.3390/jpm11111147] [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: 09/10/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
The gene coding for histone methyltransferase KMT2D is found among the top mutated genes in upper tract urothelial carcinoma (UTUC); however, there is a lack of data regarding its association with clinicopathologic features as well as survival outcomes. Therefore, we aimed to investigate KMT2D expression, mutation patterns, and their utility as prognostic biomarkers in patients with UTUC. A single-center study was conducted on tumor specimens from 51 patients treated with radical nephroureterectomy (RNU). Analysis of KMT2D protein expression was performed using immunohistochemistry (IHC). Customized next-generation sequencing (NGS) was used to assess alterations in KMT2D exons. Cox regression was used to assess the relationship of KMT2D protein expression and mutational status with survival outcomes. KMT2D expression was increased in patients with a previous history of bladder cancer (25% vs. 0%, p = 0.02). The NGS analysis of KMT2D exons in 27 UTUC tumors revealed a significant association between pathogenic KMT2D variants and tumor location (p = 0.02). Pathogenic KMT2D variants were predominantly found in patients with non-pelvic or multifocal tumors (60% vs. 14%), while the majority of patients with a pelvic tumor location (81% vs. 20%) did not harbor pathogenic KMT2D alterations. Both IHC and NGS analyses of KMT2D failed to detect a statistically significant association between KMT2D protein or KMT2D gene alteration status and clinical variables such as stage/grade of the disease or survival outcomes (all p > 0.05). KMT2D alterations and protein expression were associated with UTUC features such as multifocality, ureteral location, and previous bladder cancer. While KMT2D protein expression and KMT2D mutational status do not seem to have prognostic value in UTUC, they appear to add information to improve clinical decision-making regarding the type of therapy.
Collapse
Affiliation(s)
- Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
- Institute for Urology and Reproductive Health, Sechenov University, 119435 Moscow, Russia;
| | - Ursula Lemberger
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - Andreas Bruchbacher
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - Dafina Ilijazi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - Stephan Korn
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - Florian Berndl
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - David D’Andrea
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| | - Martin Susani
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (M.S.); (E.C.)
| | - Dmitry Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, 119435 Moscow, Russia;
| | - Eva Compérat
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (M.S.); (E.C.)
| | - Shahrokh F. Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
- Institute for Urology and Reproductive Health, Sechenov University, 119435 Moscow, Russia;
- Karl Landsteiner Institute of Urology and Andrology, 1010 Vienna, Austria
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic
| | - Melanie R. Hassler
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (E.L.); (U.L.); (A.B.); (D.I.); (S.K.); (F.B.); (D.D.); (S.F.S.)
| |
Collapse
|