1
|
Jasmin MDC, Radhakrishnan N, Prabhu VV, Sakthivel KM, Radha RR. Histone-Lysine N-Methyltransferase 2D (KMT2D) Impending Therapeutic Target for the Management of Cancer: The Giant Rats Tail. J Environ Pathol Toxicol Oncol 2025; 44:31-36. [PMID: 39462447 DOI: 10.1615/jenvironpatholtoxicoloncol.2024053872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
The histone-lysine N-methyltransferase 2D (KMT2D), tumor suppressor gene which is the major component of histone H3K4 mono-methyltransferase in mammals and has significant role in regulation of a gene which are frequently mutated that lead to many different types of cancers that include non-Hodgkin lymphoma, medulloblastoma, prostate carcinoma, renal carcinoma, bladder carcinoma and lung carcinoma. KMT2D gene epigenetic alterations in histone methylation play a significant role for the initiation and progression of cancers from pre-cancerous lesions, yet its complete function in oncogenesis remains unsolved. KMT2D deficiency - loss are thought of initial mediators of cancer development and cell migration such as B-cell lymphoma, medulloblastoma, melanoma, pancreas and lung cancer. The KMT2D loss has know to activate glycolytic genes that promote aggressive tumor progression. Therefore, the present review serves to underline the update on recent research pertaining to KMT2D gene, that could be a potential therapeutic target in downregulating glycolytic genes such as Pgk1, Ldha, Pgam1 and Gapdh; 2, epidermal growth factor receptor tyrosine kinase (EGFR-TK ) - ERBB2, RTK-RAS signaling, RAS activator genes Rgl1, Rasgrp1, Rasgrf1, Rasgrf 2 and Rapgef5 in suppressing the tumor progression that may represent novel targeted therapy for the management of cancer. This review will facilitate to understand the gene expression that inhibits cancer progression and which could serve as a potential molecular target in understanding cancer pathogenesis.
Collapse
Affiliation(s)
- Meshak Dhanashekaran Cecileya Jasmin
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Thandalam, Chennai, Tamil Nadu 602105, India; Department of Biochemistry, Tagore Medical College and Hospital, Rathinamangalam, Melakottaiyur, Chennai, Tamil Nadu 600127, India
| | | | - Venugopal Vinod Prabhu
- Department of Biochemistry, University of Madras, Guindy campus, Chennai-600025, Tamil Nadu, India
| | - Kunnathur Murugesan Sakthivel
- Karunya University, Department of Biotechnology, Karunya Nagar, Coimbatore-641114, Tamil Nadu, India; Laboratory of Cytogenetics and Molecular Diagnostics, Regional Cancer Centre, Medical College Post, Trivandrum 695011, Kerala, India
| | - Rasmi Rajan Radha
- Department of Biotechnology, PSG College of Arts & Science, Coimbatore, Tamil Nadu 614014, India
| |
Collapse
|
2
|
Gao Y, Hao J, Zhang Z. Adjuvant Chemotherapy for Breast Cancer in Older Adult Patients. Clin Interv Aging 2024; 19:1281-1286. [PMID: 39050516 PMCID: PMC11268672 DOI: 10.2147/cia.s470262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Decision-making regarding adjuvant chemotherapy for older adults with breast cancer is a challenge because older adult patients often have poor physical health, frailty, and age-related comorbidities, which can compromise treatment outcome. Due to these considerations, doctors tend to use less chemotherapy for breast cancer in older adults. However, older patients in good general health could still benefit from chemotherapy. Careful benefit-risk assessment is essential to provide best care for each older adult patient. Due to a rapidly aging population, breast cancer in older adults is becoming a serious public health issue in China. In this mini review, we discuss the need, means, and tools to assess the benefits and risks of adjuvant chemotherapy in older adults with breast cancer. The contents of this review may drive decision-making with regard to the use and selection of adjuvant chemotherapy for older adult patients in China who are fit for the treatment.
Collapse
Affiliation(s)
- Ying Gao
- Department of Breast and Thyroid Surgery, Tianjin Union Medical Center, Tianjin, 300121, People’s Republic of China
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Hospital, National Cancer Clinical Research Center, Tianjin Cancer Clinical Research Center, Tianjin Key Laboratory of Cancer Prevention and Treatment, Tianjin, 300060, People’s Republic of China
| | - Jie Hao
- Department of Breast and Thyroid Surgery, Tianjin Union Medical Center, Tianjin, 300121, People’s Republic of China
| | - Zhendong Zhang
- Department of Breast and Thyroid Surgery, Tianjin Union Medical Center, Tianjin, 300121, People’s Republic of China
| |
Collapse
|
3
|
Zhang Z, Guo Y, Gao X, Wang X, Jin C. Role of histone methyltransferase KMT2D in BMSC osteogenesis via AKT signaling. Regen Ther 2024; 26:775-782. [PMID: 39309396 PMCID: PMC11414574 DOI: 10.1016/j.reth.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Understanding the precise mechanism of BMSC (bone marrow mesenchymal stem cell) osteogenesis is critical for metabolic bone diseases and bone reconstruction. The histone-lysine N-methyltransferase 2D (KMT2D) acts as an important methyltransferase related with congenital skeletal disorders, yet the function of KMT2D in osteogenesis was unclear. Here we found that KMT2D expression was decreased in BMSCs collected from ovariectomized mice. Moreover, during human BMSC differentiation under mineralization induction, the mRNA level of KMT2D was gradually elevated. After KMT2D knockdown, the in vitro osteogenic differentiation of BMSCs was inhibited, while the in vivo bone formation potential of BMSCs was attenuated. Further, in BMSCs, KMT2D knockdown reduced the level of phosphorylated protein kinase B (p-AKT). SC-79, a common activator of AKT signaling, reversed the suppressing influence of KMT2D knockdown on BMSCs differentiation towards osteoblast. These results indicate that the KMT2D-AKT pathway plays an essential role in the osteogenesis process of human BMSCs (hBMSCs), which might provide new avenues for the molecular medicine of bone diseases and regeneration.
Collapse
Affiliation(s)
- Zhichun Zhang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Yanyan Guo
- Second Clinical Division, Peking University School and Hospital of Stomatology, Beijing, 100101, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Xuejun Gao
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Xiaoyan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Chanyuan Jin
- Second Clinical Division, Peking University School and Hospital of Stomatology, Beijing, 100101, China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| |
Collapse
|
4
|
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
|
5
|
Ding Y, Zhang C, Zuo Q, Jin K, Li B. lncCPSET1 acts as a scaffold for MLL2/COMPASS to regulate Bmp4 and promote the formation of chicken primordial germ cells. Mol Genet Genomics 2024; 299:41. [PMID: 38551742 DOI: 10.1007/s00438-024-02127-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/15/2024] [Indexed: 04/02/2024]
Abstract
Primordial germ cells (PGCs) are the ancestors of female and male germ cells. Recent studies have shown that long non-coding RNA (lncRNA) and histone methylation are key epigenetic factors affecting PGC formation; however, their joint regulatory mechanisms have rarely been studied. Here, we explored the mechanism by which lncCPSET1 and H3K4me2 synergistically regulate the formation of chicken PGCs for the first time. Combined with chromatin immunoprecipitation (CHIP) sequencing and RNA-seq of PGCs transfected with the lncCPSET1 overexpression vector, GO annotation and KEGG enrichment analysis revealed that Wnt and TGF-β signaling pathways were significantly enriched, and Fzd2, Id1, Id4, and Bmp4 were identified as candidate genes. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) showed that ASH2L, DPY30, WDR5, and RBBP5 overexpression significantly increased the expression of Bmp4, which was up-regulated after lncCPSET1 overexpression as well. It indicated that Bmp4 is a target gene co-regulated by lncCPSET1 and MLL2/COMPASS. Interestingly, co-immunoprecipitation results showed that ASH2L, DPY30 and WDR5 combined and RBBP5 weakly combined with DPY30 and WDR5. lncCPSET1 overexpression significantly increased Dpy30 expression and co-immunoprecipitation showed that interference/overexpression of lncCPSET1 did not affect the binding between the proteins in the complexes, but interference with lncCPSET1 inhibited DPY30 expression, which was confirmed by RNA immunoprecipitation that lncCPSET1 binds to DPY30. Additionally, CHIP-qPCR results showed that DPY30 enriched in the Bmp4 promoter region promoted its transcription, thus promoting the formation of PGCs. This study demonstrated that lncCPSET1 and H3K4me2 synergistically promote PGC formation, providing a reference for the study of the regulatory mechanisms between lncRNA and histone methylation, as well as a molecular basis for elucidating the formation mechanism of PGCs in chickens.
Collapse
Affiliation(s)
- Ying Ding
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, 88 South University Ave, Yangzhou, Jiangsu, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Chen Zhang
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, 88 South University Ave, Yangzhou, Jiangsu, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- RNA Medicine Center, International Institutes of Medicine, Zhejiang University, Hangzhou, China
| | - Qisheng Zuo
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, 88 South University Ave, Yangzhou, Jiangsu, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Kai Jin
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, 88 South University Ave, Yangzhou, Jiangsu, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou University, Yangzhou, 225009, China
| | - Bichun Li
- Key Laboratory of Animal Genetics, Breeding and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, 88 South University Ave, Yangzhou, Jiangsu, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| |
Collapse
|
6
|
Zhang YH, Tao Q, Zhang WY, Zhao S, Liu WP, Gao LM. Histone methyltransferase KMT2D inhibits ENKTL carcinogenesis by epigenetically activating SGK1 and SOCS1. Genes Genomics 2024; 46:203-212. [PMID: 37523130 DOI: 10.1007/s13258-023-01434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Epigenetic alteration plays an essential role in the occurrence and development of extranodal natural killer/T cell lymphoma (ENKTL). Histone methyltransferase (HMT) KMT2D is an epigenetic regulator that plays different roles in different tumors, but its role and mechanism in ENKTL are still unclear. METHODS We performed immunohistochemical staining of 112 ENKTL formalin-fixed paraffin-embedded (FFPE) samples. Then, we constructed KMT2D knockdown cell lines and conducted research on cell biological behavior. Finally, to further investigate KMT2D-mediated downstream genes, ChIP-seq and ChIP -qPCR was performed. RESULTS The low expression of KMT2D was related to a decreased abundance in histone H3 lysine 4 mono- and trimethylation (H3K4me1/3). In KMT2D knockdown YT and NK-YS cells, cell proliferation was faster (P < 0.05), apoptosis was decreased (P < 0.05), the abundance of S phase cells was increased (P < 0.05), and the level of H3K4me1 was decreased. Notably, ChIP-seq revealed two crucial genes and pathways downregulated by KMT2D. CONCLUSIONS KMT2D is a tumor suppressor gene that mediates H3K4me1 and influences ENKTL proliferation and apoptosis by regulating the cell cycle. Moreover, in ENKTL, serum- and glucocorticoid-inducible kinase-1 (SGK1) and suppressor of cytokine signaling-1 (SOCS1) are downstream genes of KMT2D.
Collapse
Affiliation(s)
- Yue-Hua Zhang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Qing Tao
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Wen-Yan Zhang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Sha Zhao
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Wei-Ping Liu
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China.
| | - Li-Min Gao
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Kalinousky AJ, Luperchio TR, Schrode KM, Harris JR, Zhang L, DeLeon VB, Fahrner JA, Lauer AM, Bjornsson HT. KMT2D Deficiency Causes Sensorineural Hearing Loss in Mice and Humans. Genes (Basel) 2023; 15:48. [PMID: 38254937 PMCID: PMC10815913 DOI: 10.3390/genes15010048] [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: 12/05/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Individuals with Kabuki syndrome type 1 (KS1) often have hearing loss recognized in middle childhood. Current clinical dogma suggests that this phenotype is caused by frequent infections due to the immune deficiency in KS1 and/or secondary to structural abnormalities of the ear. To clarify some aspects of hearing loss, we collected information on hearing status from 21 individuals with KS1 and found that individuals have both sensorineural and conductive hearing loss, with the average age of presentation being 7 years. Our data suggest that while ear infections and structural abnormalities contribute to the observed hearing loss, these factors do not explain all loss. Using a KS1 mouse model, we found hearing abnormalities from hearing onset, as indicated by auditory brainstem response measurements. In contrast to mouse and human data for CHARGE syndrome, a disorder possessing overlapping clinical features with KS and a well-known cause of hearing loss and structural inner ear abnormalities, there are no apparent structural abnormalities of the cochlea in KS1 mice. The KS1 mice also display diminished distortion product otoacoustic emission levels, which suggests outer hair cell dysfunction. Combining these findings, our data suggests that KMT2D dysfunction causes sensorineural hearing loss compounded with external factors, such as infection.
Collapse
Affiliation(s)
- Allison J. Kalinousky
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
| | - Teresa R. Luperchio
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
| | - Katrina M. Schrode
- Department of Otolaryngology-Head and Neck Surgery and Center for Hearing and Balance, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (K.M.S.); (A.M.L.)
| | - Jacqueline R. Harris
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
- Department of Neurology, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Li Zhang
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
| | - Valerie B. DeLeon
- Department of Anthropology, University of Florida, Gainesville, FL 32610, USA;
| | - Jill A. Fahrner
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Amanda M. Lauer
- Department of Otolaryngology-Head and Neck Surgery and Center for Hearing and Balance, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (K.M.S.); (A.M.L.)
| | - Hans T. Bjornsson
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; (A.J.K.); (T.R.L.); (J.R.H.); (L.Z.); (J.A.F.)
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Landspitali University Hospital, 102 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| |
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
|
Zhao Z, Dai X, Jiang G, Lin F. ASH2L Controls Ureteric Bud Morphogenesis through the Regulation of RET/GFRA1 Signaling Activity in a Mouse Model. J Am Soc Nephrol 2023; 34:988-1002. [PMID: 36758123 PMCID: PMC10278782 DOI: 10.1681/asn.0000000000000099] [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: 07/30/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT Causes of congenital anomalies of the kidney and urinary tract (CAKUT) remain unclear. The authors investigated whether and how inactivation of Ash2l -which encodes a subunit of the COMPASS methyltransferase responsible for genome-wide histone H3 lysine K4 (H3K4) methylation-might contribute to CAKUT. In a mouse model, inactivation of Ash2l in the ureteric bud (UB) lineage led to CAKUT-like phenotypes. Removal of ASH2L led to deficient H3K4 trimethylation, which slowed cell proliferation at the UB tip, delaying budding and impairing branching morphogenesis. The absence of ASH2L also downregulated the expression of Ret , Gfra1 , and Wnt11 genes involved in RET/GFRA1 signaling. These findings identify ASH2L-mediated H3K4 methylation as an upstream epigenetic regulator of signaling crucial for UB morphogenesis and indicate that deficiency or dysregulation of these processes may lead to CAKUT. BACKGROUND Ureteric bud (UB) induction and branching morphogenesis are fundamental to the establishment of the renal architecture and are key determinants of nephron number. Defective UB morphogenesis could give rise to a spectrum of malformations associated with congenital anomalies of the kidney and urinary tract (CAKUT). Signaling involving glial cell line-derived neurotrophic factor and its receptor rearranged during transfection (RET) and coreceptor GFRA1 seems to be particularly important in UB development. Recent epigenome profiling studies have uncovered dynamic changes of histone H3 lysine K4 (H3K4) methylation during metanephros development, and dysregulated H3K4 methylation has been associated with a syndromic human CAKUT. METHODS To investigate whether and how inactivation of Ash2l , which encodes a subunit of the COMPASS methyltransferase responsible for genome-wide H3K4 methylation, might contribute to CAKUT, we inactivated Ash2l specifically from the UB lineage in C57BL/6 mice and examined the effects on genome-wide H3K4 methylation and metanephros development. Genes and epigenome changes potentially involved in these effects were screened using RNA-seq combined with Cleavage Under Targets and Tagmentation sequencing. RESULTS UB-specific inactivation of Ash2l caused CAKUT-like phenotypes mainly involving renal dysplasia at birth, which were associated with deficient H3K4 trimethylation. Ash2l inactivation slowed proliferation of cells at the UB tip, delaying budding and impairing UB branching morphogenesis. These effects were associated with downregulation of Ret , Gfra1 , and Wnt11 , which participate in RET/GFRA1 signaling. CONCLUSIONS These experiments identify ASH2L-dependent H3K4 methylation in the UB lineage as an upstream epigenetic regulator of RET/GFRA1 signaling in UB morphogenesis, which, if deficient, may lead to CAKUT.
Collapse
Affiliation(s)
- Ziyi Zhao
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuantong Dai
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gengru Jiang
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Centre for Rare Disease, Shanghai, China
| | - Fujun Lin
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
R I A, Vatsyayan A, Damodaran D, Sivadas A, Van der Speeten K. Multi-omics Analysis Classifies Colorectal Cancer into Distinct Methylated Immunogenic and Angiogenic Subtypes Based on Anatomical Laterality. Indian J Surg Oncol 2023; 14:209-219. [PMID: 37359923 PMCID: PMC10284779 DOI: 10.1007/s13193-023-01760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/25/2023] [Indexed: 06/28/2023] Open
Abstract
We employed supervised machine learning algorithms to a cohort of colorectal cancer patients from the NCI to differentiate and classify the heterogenous disease based on anatomical laterality and multi-omics stratification, in a first of its kind. Multi-omics integrative analysis shows distinct clustering of left and right colorectal cancer with disentangled representation of methylome and delineation of transcriptome and genome. We present novel multi-omics findings consistent with augmented hypermethylation of genes in right CRC, epigenomic biomarkers on the right in conjunction with immune-mediated pathway signatures, and lymphocytic invasion which unlocks unique therapeutic avenues. Contrarily, left CRC multi-omics signature is found to be marked by angiogenesis, cadherins, and epithelial-mesenchymal transition (EMT). An integrated multi-omics molecular signature of RNF217-AS1, hsa-miR-10b, and panel of FBX02, FBX06, FBX044, MAD2L2, and MIIP copy number altered genes have been found by the study. Overall survival analysis reveals genomic biomarkers ABCA13 and TTN in 852 LCRC cases, and SOX11 in 170 RCRC cases that predicts a significant survival benefit. Our study exemplifies the translational competence and robustness of machine learning in effective translational bridging of research and clinic. Supplementary Information The online version contains supplementary material available at 10.1007/s13193-023-01760-6.
Collapse
Affiliation(s)
- Anu R I
- Department of Cancer Biology and Therapeutics, MVR Cancer Center and Research Institute, Calicut, Kerala India
- Department of Clinical Biochemistry, MVR Cancer Center and Research Institute, Calicut, Kerala India
| | - Aastha Vatsyayan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Dileep Damodaran
- Department of Surgical Oncology, MVR Cancer Center and Research Institute, Calicut, Kerala India
| | - Ambily Sivadas
- Division of Nutrition, St. John’s Research Institute, Bangalore, India
| | - Kurt Van der Speeten
- Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium
- Faculty of Medicine and Life Sciences, BIOMED Research Institute, University Hasselt, Hasselt, Belgium
| |
Collapse
|
11
|
D'Incal CP, Van Rossem KE, De Man K, Konings A, Van Dijck A, Rizzuti L, Vitriolo A, Testa G, Gozes I, Vanden Berghe W, Kooy RF. Chromatin remodeler Activity-Dependent Neuroprotective Protein (ADNP) contributes to syndromic autism. Clin Epigenetics 2023; 15:45. [PMID: 36945042 PMCID: PMC10031977 DOI: 10.1186/s13148-023-01450-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/16/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Individuals affected with autism often suffer additional co-morbidities such as intellectual disability. The genes contributing to autism cluster on a relatively limited number of cellular pathways, including chromatin remodeling. However, limited information is available on how mutations in single genes can result in such pleiotropic clinical features in affected individuals. In this review, we summarize available information on one of the most frequently mutated genes in syndromic autism the Activity-Dependent Neuroprotective Protein (ADNP). RESULTS Heterozygous and predicted loss-of-function ADNP mutations in individuals inevitably result in the clinical presentation with the Helsmoortel-Van der Aa syndrome, a frequent form of syndromic autism. ADNP, a zinc finger DNA-binding protein has a role in chromatin remodeling: The protein is associated with the pericentromeric protein HP1, the SWI/SNF core complex protein BRG1, and other members of this chromatin remodeling complex and, in murine stem cells, with the chromodomain helicase CHD4 in a ChAHP complex. ADNP has recently been shown to possess R-loop processing activity. In addition, many additional functions, for instance, in association with cytoskeletal proteins have been linked to ADNP. CONCLUSIONS We here present an integrated evaluation of all current aspects of gene function and evaluate how abnormalities in chromatin remodeling might relate to the pleiotropic clinical presentation in individual"s" with Helsmoortel-Van der Aa syndrome.
Collapse
Affiliation(s)
- Claudio Peter D'Incal
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Kirsten Esther Van Rossem
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
| | - Kevin De Man
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Anthony Konings
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Anke Van Dijck
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium
| | - Ludovico Rizzuti
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Alessandro Vitriolo
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Giuseppe Testa
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Human Technopole, V. Le Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Sackler School of Medicine, 727, 69978, Tel Aviv, Israel
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Epigenetic Signaling Lab (PPES), Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, 2650, Edegem, Belgium.
| |
Collapse
|
12
|
Skitchenko R, Dinikina Y, Smirnov S, Krapivin M, Smirnova A, Morgacheva D, Artomov M. Case report: Somatic mutations in microtubule dynamics-associated genes in patients with WNT-medulloblastoma tumors. Front Oncol 2023; 12:1085947. [PMID: 36713498 PMCID: PMC9877404 DOI: 10.3389/fonc.2022.1085947] [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: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor which accounts for about 20% of all pediatric brain tumors and 63% of intracranial embryonal tumors. MB is considered to arise from precursor cell populations present during an early brain development. Most cases (~70%) of MB occur at the age of 1-4 and 5-9, but are also infrequently found in adults. Total annual frequency of pediatric tumors is about 5 cases per 1 million children. WNT-subtype of MB is characterized by a high probability of remission, with a long-term survival rate of about 90%. However, in some rare cases there may be increased metastatic activity, which dramatically reduces the likelihood of a favorable outcome. Here we report two cases of MB with a histological pattern consistent with desmoplastic/nodular (DP) and classic MB, and genetically classified as WNT-MB. Both cases showed putative causal somatic protein truncating mutations identified in microtubule-associated genes: ARID2, TUBB4A, and ANK3.
Collapse
Affiliation(s)
- Rostislav Skitchenko
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia
| | - Yulia Dinikina
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Sergey Smirnov
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mikhail Krapivin
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Anna Smirnova
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Daria Morgacheva
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mykyta Artomov
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia,The Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, United States,Department of Pediatrics, Ohio State University, Columbus, OH, United States,*Correspondence: Mykyta Artomov,
| |
Collapse
|
13
|
Suter P, Dazert E, Kuipers J, Ng CKY, Boldanova T, Hall MN, Heim MH, Beerenwinkel N. Multi-omics subtyping of hepatocellular carcinoma patients using a Bayesian network mixture model. PLoS Comput Biol 2022; 18:e1009767. [PMID: 36067230 PMCID: PMC9481159 DOI: 10.1371/journal.pcbi.1009767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/16/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Comprehensive molecular characterization of cancer subtypes is essential for predicting clinical outcomes and searching for personalized treatments. We present bnClustOmics, a statistical model and computational tool for multi-omics unsupervised clustering, which serves a dual purpose: Clustering patient samples based on a Bayesian network mixture model and learning the networks of omics variables representing these clusters. The discovered networks encode interactions among all omics variables and provide a molecular characterization of each patient subgroup. We conducted simulation studies that demonstrated the advantages of our approach compared to other clustering methods in the case where the generative model is a mixture of Bayesian networks. We applied bnClustOmics to a hepatocellular carcinoma (HCC) dataset comprising genome (mutation and copy number), transcriptome, proteome, and phosphoproteome data. We identified three main HCC subtypes together with molecular characteristics, some of which are associated with survival even when adjusting for the clinical stage. Cluster-specific networks shed light on the links between genotypes and molecular phenotypes of samples within their respective clusters and suggest targets for personalized treatments.
Collapse
Affiliation(s)
- Polina Suter
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, Basel, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Charlotte K. Y. Ng
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tuyana Boldanova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Markus H. Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
14
|
Kumar J, Alspach AE, Zucker JP, Gratzinger D. Systemic Presentation of Somatic TET2 Mutated B-Cell Lymphoma in a Child With Kabuki Syndrome and a Germline KMT2D Variant. Am J Clin Pathol 2022; 158:564-569. [PMID: 36048893 DOI: 10.1093/ajcp/aqac104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Kabuki syndrome (KS) is a rare congenital malformation syndrome associated with germline KMT2D mutations. Recurrent somatic mutations in KMT2D have frequently been observed in B-cell lymphoma, but limited studies are available that evaluate the genetic landscape of B-cell lymphomas in the setting of KS. METHODS We describe a unique case of B-cell lymphoma that illustrates histologic features of pediatric-type follicular lymphoma (FL) in a young patient with KS and autoimmune disease who showed a systemic presentation of widespread lymphadenopathy and clonal lymphocytosis. RESULTS We present the first reported case of a young patient with KS harboring a germline KMT2D variant and presenting with a systemic CD10-positive, BCL2-negative B-cell lymphoma of follicle center origin illustrating histologic features of pediatric-type FL. Targeted next-generation sequencing of the B-cell lymphoma showed somatic TET2 and subclonal CXCR4 variants. These findings suggest that abnormal epigenetic regulation caused by alterations in KMT2D and TET2 may have played critical roles in promoting lymphomagenesis in this patient. CONCLUSIONS This unique case presentation highlights the importance of close clinical monitoring and the value of clinical context in the diagnosis of pediatric FL-like lesions in patients with KS.
Collapse
Affiliation(s)
- Jyoti Kumar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jacob P Zucker
- Department of Hematology/Oncology, Renown Children's Hospital, Reno, NV, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Oryani MA, Tavasoli A, Ghalavand MA, Ashtiani RZ, Rezaee A, Mahmoudi R, Golvari H, Owrangi S, Soleymani-Goloujeh M. Epigenetics and its therapeutic potential in colorectal cancer. Epigenomics 2022; 14:683-697. [PMID: 35473313 DOI: 10.2217/epi-2022-0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
It is estimated that colorectal cancer (CRC) is the leading cause of cancer-related death around the globe. 'Epigenetics' refers to changes in the chromosome rather than the DNA sequence, which may be transmitted down to daughter cells. Epigenetics is an essential part of controlling the development and variation of a single cell. ncRNAs have a role in epigenetic regulation in CRC, which will be discussed in this review in the context of DNA methylation and histone modifications. A greater survival rate for CRC patients might be achieved by addressing epigenetic mediators, as the authors show. In this review, they aim to thoroughly examine the role of epigenetics in the prognosis, diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsaneh Tavasoli
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Amin Ghalavand
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Alisam Rezaee
- Faculty of Medical Sciences & Technologies, Science & Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Hossein Golvari
- School of Nursing & Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soroor Owrangi
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Mehdi Soleymani-Goloujeh
- Department of Stem Cells & Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, ACECR, Tehran, Iran
| |
Collapse
|
16
|
Zhang ZL, Yu PF, Ling ZQ. The role of KMT2 gene in human tumors. Histol Histopathol 2022; 37:323-334. [PMID: 35233758 DOI: 10.14670/hh-18-447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Histone methylation plays a crucial role in the regulation of gene transcriptional expression, and aberration of methylation-modifying enzyme genes can lead to a variety of genetic diseases, including human cancers. The histone modified protein KMT2 (lysin methyltransferase) family are involved in cell proliferation, growth, development and differentiation through regulating gene expression, and are closely related with many blood cancers and solid tumors. In recent years, several studies have shown that mutations in the KMT2 gene occur frequently in a variety of human cancers and the mutation status of the KMT2 gene may be correlated with the occurrence, development and prognosis of some tumors. Research uncovering the clinical characteristics and molecular mechanisms of KMT2 mutation in human tumors will be helpful for early diagnosis and prognosis of tumors as well as drug development for targeted therapies.
Collapse
Affiliation(s)
- Zhi-Long Zhang
- Zhejiang Cancer Institute (Experimental Research Center), Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, PR China.,The Second Clinical Medical College of Zhejiang Chinese Medicine University, Hangzhou, PR China
| | - Peng-Fei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, PR China.
| | - Zhi-Qiang Ling
- Zhejiang Cancer Institute (Experimental Research Center), Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, PR China.
| |
Collapse
|
17
|
Mangolini A, Rocca C, Bassi C, Ippolito C, Negrini M, Dell'Atti L, Lanza G, Gafà R, Bianchi N, Pinton P, Aguiari G. DETECTION OF DISEASE‐CAUSING MUTATIONS IN PROSTATE CANCER BY NGS SEQUENCING. Cell Biol Int 2022; 46:1047-1061. [PMID: 35347810 PMCID: PMC9320837 DOI: 10.1002/cbin.11803] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
Gene mutations may affect the fate of many tumors including prostate cancer (PCa); therefore, the research of specific mutations associated with tumor outcomes might help the urologist to identify the best therapy for PCa patients such as surgical resection, adjuvant therapy or active surveillance. Genomic DNA (gDNA) was extracted from 48 paraffin‐embedded PCa samples and normal paired tissues. Next, gDNA was amplified and analyzed by next‐generation sequencing (NGS) using a specific gene panel for PCa. Raw data were refined to exclude false‐positive mutations; thus, variants with coverage and frequency lower than 100× and 5%, respectively were removed. Mutation significance was processed by Genomic Evolutionary Rate Profiling, ClinVar, and Varsome tools. Most of 3000 mutations (80%) were single nucleotide variants and the remaining 20% indels. After raw data elaboration, 312 variants were selected. Most mutated genes were KMT2D (26.45%), FOXA1 (16.13%), ATM (15.81%), ZFHX3 (9.35%), TP53 (8.06%), and APC (5.48%). Hot spot mutations in FOXA1, ATM, ZFHX3, SPOP, and MED12 were also found. Truncating mutations of ATM, lesions lying in hot spot regions of SPOP and FOXA1 as well as mutations of TP53 correlated with poor prognosis. Importantly, we have also found some germline mutations associated with hereditary cancer‐predisposing syndrome. gDNA sequencing of 48 cancer tissues by NGS allowed to detect new tumor variants as well as confirmed lesions in genes linked to prostate cancer. Overall, somatic and germline mutations linked to good/poor prognosis could represent new prognostic tools to improve the management of PCa patients.
Collapse
Affiliation(s)
- Alessandra Mangolini
- Department of Neuroscience and RehabilitationUniversity of Ferraravia fossato di mortara, 7444121FerraraItaly
| | - Christian Rocca
- UO Urology, St Anna Hospital, via Aldo Moro 844124FerraraItaly
| | - Cristian Bassi
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | | | - Massimo Negrini
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Lucio Dell'Atti
- Division of Urology, Department of Clinical, Special and Dental Science, University Hospital "Ospedali Riuniti", Marche Polytechnic University, 71 Conca Street60126AnconaItaly
| | - Giovanni Lanza
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Roberta Gafà
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Nicoletta Bianchi
- Department of Translational MedicineUniversity of Ferraravia Luigi Borsari 4644121FerraraItaly
| | - Paolo Pinton
- Department of Medical SciencesUniversity of Ferraravia fossato di mortara, 64/B44121FerraraItaly
| | - Gianluca Aguiari
- Department of Neuroscience and RehabilitationUniversity of Ferraravia fossato di mortara, 7444121FerraraItaly
| |
Collapse
|
18
|
Lu M, Li J, Fan X, Xie F, Fan J, Xiong Y. Novel Immune-Related Ferroptosis Signature in Esophageal Cancer: An Informatics Exploration of Biological Processes Related to the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 Regulatory Network. Front Genet 2022; 13:829384. [PMID: 35281840 PMCID: PMC8908453 DOI: 10.3389/fgene.2022.829384] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Considering the role of immunity and ferroptosis in the invasion, proliferation and treatment of cancer, it is of interest to construct a model of prognostic-related differential expressed immune-related ferroptosis genes (PR-DE-IRFeGs), and explore the ferroptosis-related biological processes in esophageal cancer (ESCA).Methods: Four ESCA datasets were used to identify three PR-DE-IRFeGs for constructing the prognostic model. Validation of our model was based on analyses of internal and external data sets, and comparisons with past models. With the biological-based enrichment analysis as a guide, exploration for ESCA-related biological processes was undertaken with respect to the immune microenvironment, mutations, competing endogenous RNAs (ceRNA), and copy number variation (CNV). The model’s clinical applicability was measured by nomogram and correlation analysis between risk score and gene expression, and also immune-based and chemotherapeutic sensitivity.Results: Three PR-DE-IRFeGs (DDIT3, SLC2A3, and GCH1), risk factors for prognosis of ESCA patients, were the basis for constructing the prognostic model. Validation of our model shows a meaningful capability for prognosis prediction. Furthermore, many biological functions and pathways related to immunity and ferroptosis were enriched in the high-risk group, and the role of the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 network in ESCA is supported. Also, the KMT2D mutation is associated with our risk score and SLC2A3 expression. Overall, the prognostic model was associated with treatment sensitivity and levels of gene expression.Conclusion: A novel, prognostic model was shown to have high predictive value. Biological processes related to immune functions, KMT2D mutation, CNV and the TMEM161B-AS1/hsa-miR-27a-3p/GCH1 network were involved in ESCA progression.
Collapse
Affiliation(s)
- Min Lu
- Department of Emergency, Shangrao People’s Hospital, Shangrao Hospital Affiliated to Nanchang University, Shangrao, China
| | - Jiaqi Li
- School of Stomatology, Nanchang University, Nanchang, China
| | - Xin Fan
- School of Stomatology, Nanchang University, Nanchang, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xin Fan,
| | - Fei Xie
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jie Fan
- Shangrao Municipal Hospital, Shangrao, China
| | - Yuanping Xiong
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Bousali M, Karamitros T. Hepatitis B Virus Integration into Transcriptionally Active Loci and HBV-Associated Hepatocellular Carcinoma. Microorganisms 2022; 10:microorganisms10020253. [PMID: 35208708 PMCID: PMC8879149 DOI: 10.3390/microorganisms10020253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/10/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatitis B Virus (HBV) DNA integrations into the human genome are considered major causative factors to HBV-associated hepatocellular carcinoma development. In the present study, we investigated whether HBV preferentially integrates parts of its genome in specific genes and evaluated the contribution of the integrations in HCC development per gene. We applied dedicated in-house developed pipelines on all of the available HBV DNA integration data and performed a statistical analysis to identify genes that could be characterized as hotspots of integrations, along with the evaluation of their association with HBV-HCC. Our results suggest that 15 genes are recurrently affected by HBV integrations and they are significantly associated with HBV-HCC. Further studies that focus on HBV integrations disrupting these genes are mandatory in order to understand the role of HBV integrations in clonal advantage gain and oncogenesis promotion, as well as to determine whether inhibition of the HBV-disrupted genes can provide a therapy strategy for HBV-HCC.
Collapse
Affiliation(s)
- Maria Bousali
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Laboratory of Medical Microbiology, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
- Correspondence: ; Tel.: +30-210-6478871
| |
Collapse
|
20
|
Zou B, Guo D, Kong P, Wang Y, Cheng X, Cui Y. Integrative Genomic Analyses of 1,145 Patient Samples Reveal New Biomarkers in Esophageal Squamous Cell Carcinoma. Front Mol Biosci 2022; 8:792779. [PMID: 35127817 PMCID: PMC8814608 DOI: 10.3389/fmolb.2021.792779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Due to the lack of effective diagnostic markers and therapeutic targets, esophageal squamous cell carcinoma (ESCC) shows a poor 5 years survival rate of less than 30%. To explore the potential therapeutic targets of ESCC, we integrated and reanalyzed the mutation data of WGS (whole genome sequencing) or WES (whole exome sequencing) from a total of 1,145 samples in 7 large ESCC cohorts, including 270 ESCC gene expression data. Two new mutation signatures and 20 driver genes were identified in our study. Among them, AP3S1, MUC16, and RPS15 were reported for the first time. We also discovered that the KMT2D was associated with the multiple clinical characteristics of ESCC, and KMT2D knockdown cells showed enhanced cell migration and cell invasion. Furthermore, a few neoantigens were shared between ESCC patients. For ESCC, compared to TMB, neoantigen might be treated as a better immunotherapy biomarker. Our research expands the understanding of ESCC mutations and helps the identification of ESCC biomarkers, especially for immunotherapy biomarkers.
Collapse
Affiliation(s)
- Binbin Zou
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Dinghe Guo
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Pengzhou Kong
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Yanqiang Wang
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Xiaolong Cheng
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
- *Correspondence: Xiaolong Cheng, ; Yongping Cui,
| | - Yongping Cui
- Key Laboratory of Cellular Physiology of the Ministry of Education, Shanxi Medical University, Taiyuan, China
- Department of Pathology, Shanxi Medical University, Taiyuan, China
- Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Xiaolong Cheng, ; Yongping Cui,
| |
Collapse
|
21
|
Singh AN, Sharma N. In-silico identification of frequently mutated genes and their co-enriched metabolic pathways associated with Prostate cancer progression. Andrologia 2021; 53:e14236. [PMID: 34468989 DOI: 10.1111/and.14236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/04/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022] Open
Abstract
Prostate cancer (PCa) has emerged as a significant health burden in men globally. Several genetic anomalies such as mutations and also epigenetic aberrations are responsible for the heterogeneity of this disease. This study identified the 20 most frequently mutated genes reported in PCa based on literature and database survey. Further gene ontology and functional enrichment analysis were conducted to determine their co-modulated molecular and biological pathways. A protein-protein interaction network was used for the identification of hub genes. These hub genes identified were then subjected to survival analysis. The prognostic values of these identified genes were investigated using GEPIA and HPA. Gene Ontology analysis of the identified genes depicted that these genes significantly contributed to the cell cycle, apoptosis, angiogenesis and TGF-β receptor signalling. Further, the research showed that high expressions of identified mutated genes led to a reduction in the long-term survival of PCa patients, which was supported by immunohistochemical and mRNA expression level data. Our results suggest that identified panel of mutated genes viz., CTNNB1, TP53, ATM, AR and KMT2D play crucial roles in the onset and progression of PCa, thereby providing candidate diagnostic markers for PCa for individualised treatment in the future.
Collapse
Affiliation(s)
- Anshika N Singh
- School of Engineering, Ajeenkya DY Patil University (ADYPU), Pune, India
| | - Neeti Sharma
- School of Engineering, Ajeenkya DY Patil University (ADYPU), Pune, India
| |
Collapse
|
22
|
Bousali M, Papatheodoridis G, Paraskevis D, Karamitros T. Hepatitis B Virus DNA Integration, Chronic Infections and Hepatocellular Carcinoma. Microorganisms 2021; 9:1787. [PMID: 34442866 PMCID: PMC8398950 DOI: 10.3390/microorganisms9081787] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/16/2022] Open
Abstract
Hepatitis B Virus (HBV) is an Old World virus with a high mutation rate, which puts its origins in Africa alongside the origins of Homo sapiens, and is a member of the Hepadnaviridae family that is characterized by a unique viral replication cycle. It targets human hepatocytes and can lead to chronic HBV infection either after acute infection via horizontal transmission usually during infancy or childhood or via maternal-fetal transmission. HBV has been found in ~85% of HBV-related Hepatocellular Carcinomas (HCC), and it can integrate the whole or part of its genome into the host genomic DNA. The molecular mechanisms involved in the HBV DNA integration is not yet clear; thus, multiple models have been described with respect to either the relaxed-circular DNA (rcDNA) or the double-stranded linear DNA (dslDNA) of HBV. Various genes have been found to be affected by HBV DNA integration, including cell-proliferation-related genes, oncogenes and long non-coding RNA genes (lincRNAs). The present review summarizes the advances in the research of HBV DNA integration, focusing on the evolutionary and molecular side of the integration events along with the arising clinical aspects in the light of WHO's commitment to eliminate HBV and viral hepatitis by 2030.
Collapse
Affiliation(s)
- Maria Bousali
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - George Papatheodoridis
- Department of Gastroenterology, “Laiko” General Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitrios Paraskevis
- Department of Hygiene Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Laboratory of Medical Microbiology, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
23
|
The MLL3/4 H3K4 methyltransferase complex in establishing an active enhancer landscape. Biochem Soc Trans 2021; 49:1041-1054. [PMID: 34156443 PMCID: PMC8286814 DOI: 10.1042/bst20191164] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
Enhancers are cis-regulatory elements that play essential roles in tissue-specific gene expression during development. Enhancer function in the expression of developmental genes requires precise regulation, while deregulation of enhancer function could be the main cause of tissue-specific cancer development. MLL3/KMT2C and MLL4/KMT2D are two paralogous histone modifiers that belong to the SET1/MLL (also named COMPASS) family of lysine methyltransferases and play critical roles in enhancer-regulated gene activation. Importantly, large-scale DNA sequencing studies have revealed that they are amongst the most frequently mutated genes associated with human cancers. MLL3 and MLL4 form identical multi-protein complexes for modifying mono-methylation of histone H3 lysine 4 (H3K4) at enhancers, which together with the p300/CBP-mediated H3K27 acetylation can generate an active enhancer landscape for long-range target gene activation. Recent studies have provided a better understanding of the possible mechanisms underlying the roles of MLL3/MLL4 complexes in enhancer regulation. Moreover, accumulating studies offer new insights into our knowledge of the potential role of MLL3/MLL4 in cancer development. In this review, we summarize recent evidence on the molecular mechanisms of MLL3/MLL4 in the regulation of active enhancer landscape and long-range gene expression, and discuss their clinical implications in human cancers.
Collapse
|
24
|
Dhar SS, Lee MG. Cancer-epigenetic function of the histone methyltransferase KMT2D and therapeutic opportunities for the treatment of KMT2D-deficient tumors. Oncotarget 2021; 12:1296-1308. [PMID: 34194626 PMCID: PMC8238240 DOI: 10.18632/oncotarget.27988] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/27/2022] Open
Abstract
Epigenetic mechanisms are central to understanding the molecular basis underlying tumorigenesis. Aberrations in epigenetic modifiers alter epigenomic landscapes and play a critical role in tumorigenesis. Notably, the histone lysine methyltransferase KMT2D (a COMPASS/ Set1 family member; also known as MLL4, ALR, and MLL2) is among the most frequently mutated genes in many different types of cancer. Recent studies have demonstrated how KMT2D loss induces abnormal epigenomic reprograming and rewires molecular pathways during tumorigenesis. These findings also have clinical and therapeutic implications for cancer treatment. In this review, we summarize recent advances in understanding the role of KMT2D in regulating tumorigenesis and discuss therapeutic opportunities for the treatment of KMT2D-deficient tumors.
Collapse
Affiliation(s)
- Shilpa S Dhar
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Gyu Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,The Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
25
|
Puvvula PK, Yu Y, Sullivan KR, Eyob H, Rosenberg J, Welm A, Huff C, Moon AM. Inhibiting an RBM39/MLL1 epigenomic regulatory complex with dominant-negative peptides disrupts cancer cell transcription and proliferation. Cell Rep 2021; 35:109156. [PMID: 34077726 DOI: 10.1016/j.celrep.2021.109156] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 02/07/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
RBM39 is a known splicing factor and coactivator. Here, we report that RBM39 functions as a master transcriptional regulator that interacts with the MLL1 complex to facilitate chromatin binding and H3K4 trimethylation in breast cancer cells. We identify RBM39 functional domains required for DNA and complex binding and show that the loss of RBM39 has widespread effects on H3K4me3 and gene expression, including key oncogenic pathways. RBM39's RNA recognition motif 3 (RRM3) functions as a dominant-negative domain; namely, it disrupts the complex and H3K4me trimethylation and expression of RBM/MLL1 target genes. RRM3-derived cell-penetrating peptides phenocopy the effects of the loss of RBM39 to decrease growth and survival of all major subtypes of breast cancer and yet are nontoxic to normal cells. These findings establish RBM39/MLL1 as a major contributor to the abnormal epigenetic landscape in breast cancer and lay the foundation for peptide-mediated cancer-specific therapy based on disruption of RBM39 epigenomic functions.
Collapse
Affiliation(s)
- Pavan Kumar Puvvula
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA.
| | - Yao Yu
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, TX, USA
| | - Kaelan Renaldo Sullivan
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
| | - Henok Eyob
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Julian Rosenberg
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
| | - Alana Welm
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Chad Huff
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, TX, USA
| | - Anne M Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA; Department Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
26
|
Castro-Piedras I, Sharma M, Brelsfoard J, Vartak D, Martinez EG, Rivera C, Molehin D, Bright RK, Fokar M, Guindon J, Pruitt K. Nuclear Dishevelled targets gene regulatory regions and promotes tumor growth. EMBO Rep 2021; 22:e50600. [PMID: 33860601 DOI: 10.15252/embr.202050600] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
Dishevelled (DVL) critically regulates Wnt signaling and contributes to a wide spectrum of diseases and is important in normal and pathophysiological settings. However, how it mediates diverse cellular functions remains poorly understood. Recent discoveries have revealed that constitutive Wnt pathway activation contributes to breast cancer malignancy, but the mechanisms by which this occurs are unknown and very few studies have examined the nuclear role of DVL. Here, we have performed DVL3 ChIP-seq analyses and identify novel target genes bound by DVL3. We show that DVL3 depletion alters KMT2D binding to novel targets and changes their epigenetic marks and mRNA levels. We further demonstrate that DVL3 inhibition leads to decreased tumor growth in two different breast cancer models in vivo. Our data uncover new DVL3 functions through its regulation of multiple genes involved in developmental biology, antigen presentation, metabolism, chromatin remodeling, and tumorigenesis. Overall, our study provides unique insight into the function of nuclear DVL, which helps to define its role in mediating aberrant Wnt signaling.
Collapse
Affiliation(s)
- Isabel Castro-Piedras
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Monica Sharma
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jennifer Brelsfoard
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - David Vartak
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edgar G Martinez
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Cristian Rivera
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Deborah Molehin
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Robert K Bright
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mohamed Fokar
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, USA
| | - Josee Guindon
- Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kevin Pruitt
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
27
|
Li Q, Wu R, Wu F, Chen Q. KMT2D promotes proliferation of gastric cancer cells: evidence from ctDNA sequencing. J Clin Lab Anal 2021; 35:e23721. [PMID: 33793001 PMCID: PMC8059714 DOI: 10.1002/jcla.23721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND ctDNA sequencing could be used for early cancer screening, prognosis prediction, and medication guidance. However, data of its application in gastric cancer are still lacking. In this study, using ctDNA sequencing, we aimed to screen the mutant genes closely associated with gastric cancer and to explore the impact of these genes on gastric cancer development. METHODS ctDNA for high-throughput sequencing was obtained from gastric cancer patients, and the high-frequency mutant gene KMT2D was identified. Immunohistochemical examination was conducted to assess the expression of KMT2D in gastric cancer tissues. KMT2D knockdown was performed to establish the stably transfected gastric cancer cells. Then, CCK8, plate clone formation assay, and Transwell assay were conducted, and a subcutaneous tumor-bearing model was induced in nude mice to investigate the changes in cell proliferation and invasion capability. Transcriptome sequencing was also performed to investigate the differences in cellular gene expression. RESULTS Detection of ctDNA found 113 gastric cancer-related mutations, 11 of which are the top 20 high-frequency mutations of gastric cancer recorded by COSMIC (Catalogue of Somatic Mutations in Cancer, COSMIC). They are TP53, ARID1A, CDH1, PIK3CA, KMT2C, KMT2D, APC, SPEN, CTNNB1, SETBP1, and KMT2A. The gene closely related to the clinical characteristics of the patient is KMT2D. The high-frequency mutant gene KMT2D was identified in gastric cancer tissues. The positive rate of KMT2D expression in cancer tissues was 74.3%, which was higher than that in para-carcinoma tissues (56.8%). The knockdown of KMT2D inhibited the proliferation, invasion, and tumor formation capacity of the gastric cancer cells, causing differences in the gene expression profiles, and the expression of different functional gene clusters was up- or downregulated. CONCLUSION The findings of this study revealed that KMT2D could be an oncogene capable of promoting gastric cancer proliferation.
Collapse
Affiliation(s)
- Qiaolian Li
- Fujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Geriatric HospitalFuzhouChina
| | - Riping Wu
- Fujian Medical University Union HospitalFuzhouChina
| | - Fan Wu
- Fujian Medical University Union HospitalFuzhouChina
- Fujian Provincial Cancer HospitalFuzhouChina
| | - Qiang Chen
- Fujian Medical University Union HospitalFuzhouChina
| |
Collapse
|
28
|
Boniel S, Szymańska K, Śmigiel R, Szczałuba K. Kabuki Syndrome-Clinical Review with Molecular Aspects. Genes (Basel) 2021; 12:468. [PMID: 33805950 PMCID: PMC8064399 DOI: 10.3390/genes12040468] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Kabuki syndrome (KS) is a rare developmental disorder principally comprised of developmental delay, hypotonia and a clearly defined dysmorphism: elongation of the structures surrounding the eyes, a shortened and depressed nose, thinning of the upper lip and thickening of the lower lip, large and prominent ears, hypertrichosis and scoliosis. Other characteristics include poor physical growth, cardiac, gastrointestinal and renal anomalies as well as variable behavioral issues, including autistic features. De novo or inherited pathogenic/likely pathogenic variants in the KMT2D gene are the most common cause of KS and account for up to 75% of patients. Variants in KDM6A cause up to 5% of cases (X-linked dominant inheritance), while the etiology of about 20% of cases remains unknown. Current KS diagnostic criteria include hypotonia during infancy, developmental delay and/or intellectual disability, typical dysmorphism and confirmed pathogenic/likely pathogenic variant in KMT2D or KDM6A. Care for KS patients includes the control of physical and psychomotor development during childhood, rehabilitation and multi-specialist care. This paper reviews the current clinical knowledge, provides molecular and scientific links and sheds light on the treatment of Kabuki syndrome individuals.
Collapse
Affiliation(s)
- Snir Boniel
- Department of Medical Genetics, Medical University, Pawinskiego 3c, 02-106 Warsaw, Poland;
| | - Krystyna Szymańska
- Mossakowski Medical Research Center, Department of Experimental and Clinical Neuropathology, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Robert Śmigiel
- Department of Paediatrics, Division of Propaedeutic of Paediatrics and Rare Disorders, Medical University, 51-618 Wroclaw, Poland;
| | - Krzysztof Szczałuba
- Department of Medical Genetics, Medical University, Pawinskiego 3c, 02-106 Warsaw, Poland;
| |
Collapse
|
29
|
Zhang L, Pilarowski G, Pich EM, Nakatani A, Dunlop J, Baba R, Matsuda S, Daini M, Hattori Y, Matsumoto S, Ito M, Kimura H, Bjornsson HT. Inhibition of KDM1A activity restores adult neurogenesis and improves hippocampal memory in a mouse model of Kabuki syndrome. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:779-791. [PMID: 33738331 PMCID: PMC7940709 DOI: 10.1016/j.omtm.2021.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Kabuki syndrome (KS) is a rare cause of intellectual disability primarily caused by loss-of-function mutations in lysine-specific methyltransferase 2D (KMT2D), which normally adds methyl marks to lysine 4 on histone 3. Previous studies have shown that a mouse model of KS (Kmt2d+/βGeo) demonstrates disruption of adult neurogenesis and hippocampal memory. Proof-of-principle studies have shown postnatal rescue of neurological dysfunction following treatments that promote chromatin opening; however, these strategies are non-specific and do not directly address the primary defect of histone methylation. Since lysine-specific demethylase 1A (LSD1/KDM1A) normally removes the H3K4 methyl marks added by KMT2D, we hypothesized that inhibition of KDM1A demethylase activity may ameliorate molecular and phenotypic defects stemming from KMT2D loss. To test this hypothesis, we evaluated a recently developed KDM1A inhibitor (TAK-418) in Kmt2d+/βGeo mice. We found that orally administered TAK-418 increases the numbers of newly born doublecortin (DCX)+ cells and processes in the hippocampus in a dose-dependent manner. We also observed TAK-418-dependent rescue of histone modification defects in hippocampus both by western blot and chromatin immunoprecipitation sequencing (ChIP-seq). Treatment rescues gene expression abnormalities including those of immediate early genes such as FBJ osteosarcoma oncogene (Fos) and FBJ osteosarcoma oncogene homolog B (Fosb). After 2 weeks of TAK-418, Kmt2d+/βGeo mice demonstrated normalization of hippocampal memory defects. In summary, our data suggest that KDM1A inhibition is a plausible treatment strategy for KS and support the hypothesis that the epigenetic dysregulation secondary to KMT2D dysfunction plays a major role in the postnatal neurological disease phenotype in KS.
Collapse
Affiliation(s)
- Li Zhang
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Genay Pilarowski
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Atsushi Nakatani
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - John Dunlop
- Takeda Pharmaceutical Company Limited, London, UK
| | - Rina Baba
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Satoru Matsuda
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Masaki Daini
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yasushi Hattori
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | | | - Mitsuhiro Ito
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Haruhide Kimura
- Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Hans Tomas Bjornsson
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik 101, Iceland
- Landspitali University Hospital, Reykjavik 101, Iceland
- Corresponding author: Hans Tomas Bjornsson, McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway Street, MRB 415, Baltimore, MD 21205, USA.
| |
Collapse
|
30
|
Ferrer AI, Trinidad JR, Sandiford O, Etchegaray JP, Rameshwar P. Epigenetic dynamics in cancer stem cell dormancy. Cancer Metastasis Rev 2021; 39:721-738. [PMID: 32394305 DOI: 10.1007/s10555-020-09882-x] [Citation(s) in RCA: 22] [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] [Indexed: 02/07/2023]
Abstract
Cancer remains one of the most challenging diseases despite significant advances of early diagnosis and therapeutic treatments. Cancerous tumors are composed of various cell types including cancer stem cells capable of self-renewal, proliferation, differentiation, and invasion of distal tumor sites. Most notably, these cells can enter a dormant cellular state that is resistant to conventional therapies. Thereby, cancer stem cells have the intrinsic potential for tumor initiation, tumor growth, metastasis, and tumor relapse after therapy. Both genetic and epigenetic alterations are attributed to the formation of multiple tumor types. This review is focused on how epigenetic dynamics involving DNA methylation and DNA oxidations are implicated in breast cancer and glioblastoma multiforme. The emergence and progression of these cancer types rely on cancer stem cells with the capacity to enter quiescence also known as a dormant cellular state, which dictates the distinct tumorigenic aggressiveness between breast cancer and glioblastomas.
Collapse
Affiliation(s)
- Alejandra I Ferrer
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Jonathan R Trinidad
- Department of Biological Sciences, Rutgers University, Newark, NJ, 07102, USA
| | - Oleta Sandiford
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | | | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
31
|
Huisman C, Kim YA, Jeon S, Shin B, Choi J, Lim SJ, Youn SM, Park Y, K C M, Kim S, Lee SK, Lee S, Lee JW. The histone H3-lysine 4-methyltransferase Mll4 regulates the development of growth hormone-releasing hormone-producing neurons in the mouse hypothalamus. Nat Commun 2021; 12:256. [PMID: 33431871 PMCID: PMC7801453 DOI: 10.1038/s41467-020-20511-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
In humans, inactivating mutations in MLL4, which encodes a histone H3-lysine 4-methyltransferase, lead to Kabuki syndrome (KS). While dwarfism is a cardinal feature of KS, the underlying etiology remains unclear. Here we report that Mll4 regulates the development of growth hormone-releasing hormone (GHRH)-producing neurons in the mouse hypothalamus. Our two Mll4 mutant mouse models exhibit dwarfism phenotype and impairment of the developmental programs for GHRH-neurons. Our ChIP-seq analysis reveals that, in the developing mouse hypothalamus, Mll4 interacts with the transcription factor Nrf1 to trigger the expression of GHRH-neuronal genes. Interestingly, the deficiency of Mll4 results in a marked reduction of histone marks of active transcription, while treatment with the histone deacetylase inhibitor AR-42 rescues the histone mark signature and restores GHRH-neuronal production in Mll4 mutant mice. Our results suggest that the developmental dysregulation of Mll4-directed epigenetic control of transcription plays a role in the development of GHRH-neurons and dwarfism phenotype in mice.
Collapse
Affiliation(s)
- Christian Huisman
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Young A Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Shin Jeon
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Bongjin Shin
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Jeonghoon Choi
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Su Jeong Lim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Sung Min Youn
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Younjung Park
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Medha K C
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Sangsoo Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Soo-Kyung Lee
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea.
| | - Jae W Lee
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 142604, USA.
| |
Collapse
|
32
|
Schwenty-Lara J, Pauli S, Borchers A. Using Xenopus to analyze neurocristopathies like Kabuki syndrome. Genesis 2020; 59:e23404. [PMID: 33351273 DOI: 10.1002/dvg.23404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 11/08/2022]
Abstract
Neurocristopathies are human congenital syndromes that arise from defects in neural crest (NC) development and are typically associated with malformations of the craniofacial skeleton. Genetic analyses have been very successful in identifying pathogenic mutations, however, model organisms are required to characterize how these mutations affect embryonic development thereby leading to complex clinical conditions. The African clawed frog Xenopus laevis provides a broad range of in vivo and in vitro tools allowing for a detailed characterization of NC development. Due to the conserved nature of craniofacial morphogenesis in vertebrates, Xenopus is an efficient and versatile system to dissect the morphological and cellular phenotypes as well as the signaling events leading to NC defects. Here, we review a set of techniques and resources how Xenopus can be used as a disease model to investigate the pathogenesis of Kabuki syndrome and neurocristopathies in a wider sense.
Collapse
Affiliation(s)
- Janina Schwenty-Lara
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Marburg, Germany
| | - Silke Pauli
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Annette Borchers
- Department of Biology, Molecular Embryology, Philipps-University Marburg, Marburg, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
33
|
Yu Q, Liao Z, Liu D, Xie W, Liu Z, Liao G, Wang C. Small molecule inhibitors of the prostate cancer target KMT2D. Biochem Biophys Res Commun 2020; 533:540-547. [PMID: 32988590 DOI: 10.1016/j.bbrc.2020.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/25/2022]
Abstract
Histone lysine N-methyltransferase 2D (KMT2D), an important methyltransferase that is involved in the methylation of lysine 4 in histone H3 (H3K4) and related to the development of prostate cancer. Hypermethylation of H3K4 is shown in prostate cancer (PCa). However, KMT2D inhibitors have not yet been developed. This article aims to design small molecule inhibitors targeting KMT2D_SET to prevent PCa cell proliferation and migration. Twenty-four inhibitors were firstly designed according to a virtual screening of computers,and shown different degrees of binding to KMT2D_SET. Compounds 1 and 16 showed high binding affinities to KMT2D, with KD values of 147 ± 32.9 μM and 176 ± 37.9 μM, respectively. In addition, they exerted strong inhibitory activity against the PCa cell lines PC-3 and DU145, with IC50 values of 1.1 ± 0.06 μM, 1.5 ± 0.06 μM and 1.8 ± 0.1 μM, 2.3 ± 0.2 μM, respectively. Furthermore, these two compounds significantly suppressed the migration of PCa cells.
Collapse
Affiliation(s)
- Qi Yu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zonglang Liao
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Dan Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Wei Xie
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Guochao Liao
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Caiyan Wang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
34
|
Arora R, Choi JE, Harms PW, Chandrani P. Merkel Cell Polyomavirus in Merkel Cell Carcinoma: Integration Sites and Involvement of the KMT2D Tumor Suppressor Gene. Viruses 2020; 12:v12090966. [PMID: 32878339 PMCID: PMC7552051 DOI: 10.3390/v12090966] [Citation(s) in RCA: 4] [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: 08/05/2020] [Revised: 08/15/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Merkel cell carcinoma (MCC) is an uncommon, lethal cancer of the skin caused by either Merkel cell polyomavirus (MCPyV) or UV-linked mutations. MCPyV is found integrated into MCC tumor genomes, accompanied by truncation mutations that render the MCPyV large T antigen replication incompetent. We used the open access HPV Detector/Cancer-virus Detector tool to determine MCPyV integration sites in whole-exome sequencing data from five MCC cases, thereby adding to the limited published MCPyV integration site junction data. We also systematically reviewed published data on integration for MCPyV in the human genome, presenting a collation of 123 MCC cases and their linked chromosomal sites. We confirmed that there were no highly recurrent specific sites of integration. We found that chromosome 5 was most frequently involved in MCPyV integration and that integration sites were significantly enriched for genes with binding sites for oncogenic transcription factors such as LEF1 and ZEB1, suggesting the possibility of increased open chromatin in these gene sets. Additionally, in one case we found, for the first time, integration involving the tumor suppressor gene KMT2D, adding to previous reports of rare MCPyV integration into host tumor suppressor genes in MCC.
Collapse
MESH Headings
- Carcinoma, Merkel Cell/genetics
- Carcinoma, Merkel Cell/metabolism
- Carcinoma, Merkel Cell/virology
- Cell Line, Tumor
- Chromosomes, Human, Pair 5/genetics
- Chromosomes, Human, Pair 5/metabolism
- Chromosomes, Human, Pair 5/virology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Genes, Tumor Suppressor
- Humans
- Merkel cell polyomavirus/genetics
- Merkel cell polyomavirus/physiology
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Polyomavirus Infections/genetics
- Polyomavirus Infections/metabolism
- Polyomavirus Infections/virology
- Tumor Virus Infections/genetics
- Tumor Virus Infections/metabolism
- Tumor Virus Infections/virology
- Virus Integration
- Zinc Finger E-box-Binding Homeobox 1/genetics
- Zinc Finger E-box-Binding Homeobox 1/metabolism
Collapse
Affiliation(s)
- Reety Arora
- Cellular Organization and Signalling Group, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
- Correspondence:
| | - Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; (J.E.C.); (P.W.H.)
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- School of Medicine, University of San Diego, San Diego, CA 92093, USA
| | - Paul W. Harms
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; (J.E.C.); (P.W.H.)
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pratik Chandrani
- Medical Oncology Molecular Laboratory, Medical Oncology Department, Tata Memorial Hospital, Mumbai 400012, India;
- Centre for Computational Biology, Bioinformatics and Crosstalk Laboratory, ACTREC–Tata Memorial Centre, Navi Mumbai 410210, India
| |
Collapse
|
35
|
Cheng J, Jiang R, Yang Y, Cao X, Huang Y, Lan X, Lei C, Hu L, Chen H. Association analysis of KMT2D copy number variation as a positional candidate for growth traits. Gene 2020; 753:144799. [DOI: 10.1016/j.gene.2020.144799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/25/2022]
|
36
|
Pacelli C, Adipietro I, Malerba N, Squeo GM, Piccoli C, Amoresano A, Pinto G, Pucci P, Lee JE, Ge K, Capitanio N, Merla G. Loss of Function of the Gene Encoding the Histone Methyltransferase KMT2D Leads to Deregulation of Mitochondrial Respiration. Cells 2020; 9:cells9071685. [PMID: 32668765 PMCID: PMC7407568 DOI: 10.3390/cells9071685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
KMT2D encodes a methyltransferase responsible for histone 3 lysine 4 (H3K4) mono-/di-methylation, an epigenetic mark correlated with active transcription. Here, we tested the hypothesis that KMT2D pathogenic loss-of-function variants, which causes the Kabuki syndrome type 1, could affect the mitochondrial metabolic profile. By using Seahorse technology, we showed a significant reduction of the mitochondrial oxygen consumption rate as well as a reduction of the glycolytic flux in both Kmt2d knockout MEFs and skin fibroblasts of Kabuki patients harboring heterozygous KMT2D pathogenic variants. Mass-spectrometry analysis of intermediate metabolites confirmed alterations in the glycolytic and TCA cycle pathways. The observed metabolic phenotype was accompanied by a significant increase in the production of reactive oxygen species. Measurements of the specific activities of the mitochondrial respiratory chain complexes revealed significant inhibition of CI (NADH dehydrogenase) and CIV (cytochrome c oxidase); this result was further supported by a decrease in the protein content of both complexes. Finally, we unveiled an impaired oxidation of glucose and larger reliance on long-chain fatty acids oxidation. Altogether, our findings clearly indicate a rewiring of the mitochondrial metabolic phenotype in the KMT2D-null or loss-of-function context that might contribute to the development of Kabuki disease, and represents metabolic reprogramming as a potential new therapeutic approach.
Collapse
Affiliation(s)
- Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (C.P.)
| | - Iolanda Adipietro
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy; (I.A.); (N.M.); (G.M.S.)
| | - Natascia Malerba
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy; (I.A.); (N.M.); (G.M.S.)
| | - Gabriella Maria Squeo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy; (I.A.); (N.M.); (G.M.S.)
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (C.P.)
| | - Angela Amoresano
- Department of Chemical Sciences, University of Naples “Federico II”, 80126 Napoli, Italy; (A.A.); (G.P.); (P.P.)
- INBB—Biostructures and Biosystems National Institute, 00136 Roma, Italy
| | - Gabriella Pinto
- Department of Chemical Sciences, University of Naples “Federico II”, 80126 Napoli, Italy; (A.A.); (G.P.); (P.P.)
| | - Pietro Pucci
- Department of Chemical Sciences, University of Naples “Federico II”, 80126 Napoli, Italy; (A.A.); (G.P.); (P.P.)
- Department of Chemical Sciences, CEINGE Advanced Biotechnology, Federico II University, 80145 Napoli, Italy
| | - Ji-Eun Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (J.E.L.); (K.G.)
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (J.E.L.); (K.G.)
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (C.P.)
- Correspondence: (N.C.); (G.M.)
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy; (I.A.); (N.M.); (G.M.S.)
- Correspondence: (N.C.); (G.M.)
| |
Collapse
|
37
|
Oxidative stress abrogates the degradation of KMT2D to promote degeneration in nucleus pulposus. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165888. [PMID: 32599142 DOI: 10.1016/j.bbadis.2020.165888] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/14/2020] [Accepted: 06/23/2020] [Indexed: 01/20/2023]
Abstract
Nucleus pulposus (NP) degeneration plays pivotal roles in intervertebral disc degeneration. The effect and mechanism of oxidative stress and epigenetics in NP degeneration is still unclear. We performed this study to evaluate the function of oxidative stress in NP and to explore the potential mechanism of ROS induced expression of matrix metalloproteinases (MMPs). We tested four methyltransferases, KMT2A, KMT2B, KMT2C and KMT2D in human NP samples, only KMT2D was significantly up-regulated in the severe degeneration samples. Knockdown of Kmt2d by siRNA significantly down-regulated the expression levels of catabolic enzymes including Mmp3, Mmp9 and Mmp13. Moreover, an interaction between KMT2D and ubiquitination was confirmed, and the application of H2O2 abrogated this process. Co-IP assay confirmed that H2O2 induced the phosphorylation of KMT2D to block the ubiquitination degradation, which was mainly mediated by phosphorylation of p38/MAPK. Further investigation suggested that ROS induced the alteration in levels of methylation is linked to H3K4me1 and H3K4me2, but not me3. However, usage of OICR-9429 (OICR) also suppressed the expression levels of Mmp3, Mmp9 and Mmp13. In an ex vivo model, application of OICR-9429 (OICR) also attenuated the degeneration of NP according to the H&E and Safranin-O/Fast Green staining assay, and the protein levels of MMP3, MMP9 and MMP13 were down-regulated, as well. In conclusion, we approved that oxidative stress induced ROS production promote the process of NP degeneration by enhancing KMT2D mediated transcriptional regulation of matrix degeneration related genes during NP degeneration.
Collapse
|
38
|
Bae S, Lesch BJ. H3K4me1 Distribution Predicts Transcription State and Poising at Promoters. Front Cell Dev Biol 2020; 8:289. [PMID: 32432110 PMCID: PMC7214686 DOI: 10.3389/fcell.2020.00289] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/03/2020] [Indexed: 11/13/2022] Open
Abstract
Monomethylation on lysine 4 of histone H3 (H3K4me1) is commonly associated with distal enhancers, but H3K4me1 is also present at promoter regions proximal to transcription start sites. To assess a possible role for H3K4me1 in dictating gene regulatory states at promoters, we examined H3K4me1 peak density around promoters in human and mouse germ cells using an analytic strategy that allowed us to assess relationships between different epigenetic marks on a promoter-by-promoter basis. We found that H3K4me1 exhibits either a bimodal pattern at active promoters, where it flanks H3K4me3, or a unimodal pattern at poised promoters, where it coincides with both H3K4me3 and H3K27me3. This pattern is correlated with gene expression level, but is more strongly linked to a poised chromatin state, defined by the simultaneous presence of H3K4me3 and H3K27me3, than to transcriptional activity. The pattern is especially prominent in germ cells, but is also present in other cell types, including embryonic stem cells and differentiated somatic cells. We propose that H3K4me1 is a key feature of the poised epigenetic state, and suggest possible roles for this mark in epigenetic memory.
Collapse
Affiliation(s)
- Sunhee Bae
- Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Bluma J Lesch
- Department of Genetics, Yale School of Medicine, New Haven, CT, United States.,Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
39
|
Lavery WJ, Barski A, Wiley S, Schorry EK, Lindsley AW. KMT2C/D COMPASS complex-associated diseases [K CDCOM-ADs]: an emerging class of congenital regulopathies. Clin Epigenetics 2020; 12:10. [PMID: 31924266 PMCID: PMC6954584 DOI: 10.1186/s13148-019-0802-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022] Open
Abstract
The type 2 lysine methyltransferases KMT2C and KMT2D are large, enzymatically active scaffold proteins that form the core of nuclear regulatory structures known as KMT2C/D COMPASS complexes (complex of proteins associating with Set1). These evolutionarily conserved proteins regulate DNA promoter and enhancer elements, modulating the activity of diverse cell types critical for embryonic morphogenesis, central nervous system development, and post-natal survival. KMT2C/D COMPASS complexes and their binding partners enhance active gene expression of specific loci via the targeted modification of histone-3 tail residues, in general promoting active euchromatic conformations. Over the last 20 years, mutations in five key COMPASS complex genes have been linked to three human congenital syndromes: Kabuki syndrome (type 1 [KMT2D] and 2 [KDM6A]), Rubinstein-Taybi syndrome (type 1 [CBP] and 2 [EP300]), and Kleefstra syndrome type 2 (KMT2C). Here, we review the composition and biochemical function of the KMT2 complexes. The specific cellular and embryonic roles of the KMT2C/D COMPASS complex are highlight with a focus on clinically relevant mechanisms sensitive to haploinsufficiency. The phenotypic similarities and differences between the members of this new family of disorders are outlined and emerging therapeutic strategies are detailed.
Collapse
Affiliation(s)
- William J Lavery
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA
| | - Artem Barski
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA
- Division of Human Genetics, CCHMC, Cincinnati, OH, USA
| | - Susan Wiley
- Division of Developmental and Behavioral Pediatrics, CCHMC, Cincinnati, OH, USA
| | | | - Andrew W Lindsley
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center (CCHMC), 3333 Burnet Avenue, Cincinnati, OH, 45229-3026, USA.
| |
Collapse
|
40
|
Strub T, Ballotti R, Bertolotto C. The "ART" of Epigenetics in Melanoma: From histone "Alterations, to Resistance and Therapies". Theranostics 2020; 10:1777-1797. [PMID: 32042336 PMCID: PMC6993228 DOI: 10.7150/thno.36218] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
Malignant melanoma is the most deadly form of skin cancer. It originates from melanocytic cells and can also arise at other body sites. Early diagnosis and appropriate medical care offer excellent prognosis with up to 5-year survival rate in more than 95% of all patients. However, long-term survival rate for metastatic melanoma patients remains at only 5%. Indeed, malignant melanoma is known for its notorious resistance to most current therapies and is characterized by both genetic and epigenetic alterations. In cutaneous melanoma (CM), genetic alterations have been implicated in drug resistance, yet the main cause of this resistance seems to be non-genetic in nature with a change in transcription programs within cell subpopulations. This change can adapt and escape targeted therapy and immunotherapy cytotoxic effects favoring relapse. Because they are reversible in nature, epigenetic changes are a growing focus in cancer research aiming to prevent or revert the drug resistance with current therapies. As such, the field of epigenetic therapeutics is among the most active area of preclinical and clinical research with effects of many classes of epigenetic drugs being investigated. Here, we review the multiplicity of epigenetic alterations, mainly histone alterations and chromatin remodeling in both cutaneous and uveal melanomas, opening opportunities for further research in the field and providing clues to specifically control these modifications. We also discuss how epigenetic dysregulations may be exploited to achieve clinical benefits for the patients, the limitations of these therapies, and recent data exploring this potential through combinatorial epigenetic and traditional therapeutic approaches.
Collapse
Affiliation(s)
- Thomas Strub
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Robert Ballotti
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Corine Bertolotto
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| |
Collapse
|
41
|
Li H, Li Q, Lian J, Chu Y, Fang K, Xu A, Chen T, Xu M. MLL2 promotes cancer cell lymph node metastasis by interacting with RelA and facilitating STC1 transcription. Cell Signal 2019; 65:109457. [PMID: 31676369 DOI: 10.1016/j.cellsig.2019.109457] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) presents with lymph node metastasis in the early stages, limiting the opportunities for curative local resection, including endoscopic submucosal dissection (ESD). ESD is regarded as the standard treatment for early-stage ESCCs. However, radical surgery is recommended when lymph node metastasis risk exists. More efforts are needed to find the markers for early prediction and clarify the molecular mechanism underlying the pathogenesis of lymph node metastasis. Recently, aberrant regulation of gene expression by histone methylation modifiers has emerged as an important mechanism for cancer metastasis. Herein, we demonstrated that mixed-lineage leukemia 2 (MLL2) positively regulates gene expression programs associated with ESCC cell migration. MLL2 interacts with RelA in the nucleus to enhance transcription of stanniocalcin-1 (STC1) and to facilitate cancer metastasis. Meanwhile, MLL2 knockdown resulted in a significant decrease in the migration of ESCC cells. Clinically, high level of MLL2 was significantly associated with early-stage ESCC lymph node metastasis. In summary, these findings discovered a previously unidentified molecular pathway underlying the coordinated regulation of metastasis-related STC-1 expression by MLL2 and RelA and highlighted the critical role of MLL2 in ESCC.
Collapse
Affiliation(s)
- Hongqi Li
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinfang Li
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjing Lian
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan Chu
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kang Fang
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Aiping Xu
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Chen
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Meidong Xu
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
42
|
Md Nasir ND, Ng CCY, Rajasegaran V, Wong SF, Liu W, Ng GXP, Lee JY, Guan P, Lim JQ, Thike AA, Koh VCY, Loke BN, Chang KTE, Gudi MA, Lian DWQ, Madhukumar P, Tan BKT, Tan VKM, Wong CY, Yong WS, Ho GH, Ong KW, Tan P, Teh BT, Tan PH, Rahman NA, Nahar Begum SMK, Cheah PL, Chen CJ, Dela Fuente E, Han A, Harada O, Kanomata N, Lee CS, Han Lee JY, Kamal M, Nishimura R, Ohi Y, Sawyer EJ, Teoh KH, Tsang AKH, Tsang JY, Tse GMK, Yamaguchi R. Genomic characterisation of breast fibroepithelial lesions in an international cohort. J Pathol 2019; 249:447-460. [DOI: 10.1002/path.5333] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/11/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
|
43
|
Hou G, Xu W, Jin Y, Wu J, Pan Y, Zhou F. MiRNA-217 accelerates the proliferation and migration of bladder cancer via inhibiting KMT2D. Biochem Biophys Res Commun 2019; 519:747-753. [PMID: 31547991 DOI: 10.1016/j.bbrc.2019.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022]
Abstract
To uncover the biological function of miRNA-217 in the progression of bladder cancer and the underlying mechanism. Potential miRNAs binding KMT2D were predicted through online bioinformatics. Their expression levels in bladder cancer tissues and adjacent ones were determined. Through Pearson correlation analysis and survival analysis, the most potential miRNA candidate (miRNA-217) that targets and regulates KMT2D in bladder cancer was selected. Subsequently, expression levels of miRNA-217 and KMT2D in non-muscle invasive bladder cancer (NMIBC) and muscle invasive bladder cancer (MIBC) were detected. MiRNA-217 level in bladder cancer cell lines was determined as well. The interaction between KMT2D and miRNA-217 was verified by dual-luciferase reporter gene assay. Finally, regulatory effect of miRNA-217 on viability and migration in T24 and UMUC-3 cells were investigated. Five potential candidates that were upstream genes binding KMT2D were searched by bioinformatics. Among them, miRNA-217 was remarkably upregulated in bladder cancer tissues and closely linked to poor prognosis of affected patients. Moreover, dual-luciferase reporter gene assay verified the interaction between miRNA-217 and KMT2D. MiRNA-217 was able to downregulate mRNA and protein levels of KMT2D. Furthermore, knockdown of miRNA-217 attenuated viability and migration in bladder cancer cells. MiRNA-217 accelerates proliferative and migratory abilities in bladder cancer via inhibiting the level of tumor suppressor KMT2D, thereafter leading to the poor prognosis in bladder cancer patients.
Collapse
Affiliation(s)
- Guoliang Hou
- Department of Urology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, China; Department of Urology, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan, 528000, Guangdong, China
| | - Wenfeng Xu
- Department of Urology, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan, 528000, Guangdong, China
| | - Yabin Jin
- Clinical Research Institute, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan, 528000, Guangdong, China
| | - Jialing Wu
- Clinical Research Institute, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan, 528000, Guangdong, China
| | - Yingming Pan
- Clinical Research Institute, The First People's Hospital of Foshan (Affiliated Foshan Hospital of Sun Yat-sen University), Foshan, 528000, Guangdong, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
44
|
Grant AD, Vail P, Padi M, Witkiewicz AK, Knudsen ES. Interrogating Mutant Allele Expression via Customized Reference Genomes to Define Influential Cancer Mutations. Sci Rep 2019; 9:12766. [PMID: 31484939 PMCID: PMC6726654 DOI: 10.1038/s41598-019-48967-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022] Open
Abstract
Genetic alterations are essential for cancer initiation and progression. However, differentiating mutations that drive the tumor phenotype from mutations that do not affect tumor fitness remains a fundamental challenge in cancer biology. To better understand the impact of a given mutation within cancer, RNA-sequencing data was used to categorize mutations based on their allelic expression. For this purpose, we developed the MAXX (Mutation Allelic Expression Extractor) software, which is highly effective at delineating the allelic expression of both single nucleotide variants and small insertions and deletions. Results from MAXX demonstrated that mutations can be separated into three groups based on their expression of the mutant allele, lack of expression from both alleles, or expression of only the wild-type allele. By taking into consideration the allelic expression patterns of genes that are mutated in PDAC, it was possible to increase the sensitivity of widely used driver mutation detection methods, as well as identify subtypes that have prognostic significance and are associated with sensitivity to select classes of therapeutic agents in cell culture. Thus, differentiating mutations based on their mutant allele expression via MAXX represents a means to parse somatic variants in tumor genomes, helping to elucidate a gene’s respective role in cancer.
Collapse
Affiliation(s)
- Adam D Grant
- University of Arizona Cancer Center, Tucson, AZ, 85719, USA
| | - Paris Vail
- University of Arizona Cancer Center, Tucson, AZ, 85719, USA
| | - Megha Padi
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, 85719, USA
| | | | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
45
|
Scala M, Morana G, Sementa AR, Merla G, Piatelli G, Capra V, Pavanello M. Aggressive desmoid fibromatosis in Kabuki syndrome: Expanding the tumor spectrum. Pediatr Blood Cancer 2019; 66:e27831. [PMID: 31131956 DOI: 10.1002/pbc.27831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 05/16/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Marcello Scala
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giovanni Morana
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Gianluca Piatelli
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Valeria Capra
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Pavanello
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
46
|
Li SS, Jiang WL, Xiao WQ, Li K, Zhang YF, Guo XY, Dai YQ, Zhao QY, Jiang MJ, Lu ZJ, Wan R. KMT2D deficiency enhances the anti-cancer activity of L48H37 in pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2019; 11:599-621. [PMID: 31435462 PMCID: PMC6700028 DOI: 10.4251/wjgo.v11.i8.599] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/23/2019] [Accepted: 02/27/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Novel therapeutic strategies are urgently needed for patients with a delayed diagnosis of pancreatic ductal adenocarcinoma (PDAC) in order to improve their chances of survival. Recent studies have shown potent anti-neoplastic effects of curcumin and its analogues. In addition, the role of histone methyltransferases on cancer therapeutics has also been elucidated. However, the relationship between these two factors in the treatment of pancreatic cancer remains unknown. Our working hypothesis was that L48H37, a novel curcumin analog, has better efficacy in pancreatic cancer cell growth inhibition in the absence of histone-lysine N-methyltransferase 2D (KMT2D).
AIM To determine the anti-cancer effects of L48H37 in PDAC, and the role of KMT2D on its therapeutic efficacy.
METHODS The viability and proliferation of primary (PANC-1 and MIA PaCa-2) and metastatic (SW1990 and ASPC-1) PDAC cell lines treated with L48H37 was determined by CCK8 and colony formation assay. Apoptosis, mitochondrial membrane potential (MMP), reactive oxygen species (ROS) levels, and cell cycle profile were determined by staining the cells with Annexin-V/7-AAD, JC-1, DCFH-DA, and PI respectively, as well as flow cytometric acquisition. In vitro migration was assessed by the wound healing assay. The protein and mRNA levels of relevant factors were analyzed using Western blotting, immunofluorescence and real time-quantitative PCR. The in situ expression of KMT2D in both human PDAC and paired adjacent normal tissues was determined by immunohistochemistry. In vivo tumor xenografts were established by injecting nude mice with PDAC cells. Bioinformatics analyses were also conducted using gene expression databases and TCGA.
RESULTS L48H37 inhibited the proliferation and induced apoptosis in SW1990 and ASPC-1 cells in a dose- and time-dependent manner, while also reducing MMP, increasing ROS levels, arresting cell cycle at the G2/M stages and activating the endoplasmic reticulum (ER) stress-associated protein kinase RNA-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 (ATF4)/CHOP signaling pathway. Knocking down ATF4 significantly upregulated KMT2D in PDAC cells, and also decreased L48H37-induced apoptosis. Furthermore, silencing KMT2D in L48H37-treated cells significantly augmented apoptosis and the ER stress pathway, indicating that KMT2D depletion is essential for the anti-neoplastic effects of L48H37. Administering L48H37 to mice bearing tumors derived from control or KMT2D-knockdown PDAC cells significantly decreased the tumor burden. We also identified several differentially expressed genes in PDAC cell lines expressing very low levels of KMT2D that were functionally categorized into the extrinsic apoptotic signaling pathway. The KMT2D high- and low-expressing PDAC patients from the TCGA database showed similar survival rates,but higher KMT2D expression was associated with poor tumor grade in clinical and pathological analyses.
CONCLUSION L48H37 exerts a potent anti-cancer effect in PDAC, which is augmented by KMT2D deficiency.
Collapse
Affiliation(s)
- Si-Si Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Wei-Liang Jiang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Wen-Qin Xiao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Kai Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Ye-Fei Zhang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Xing-Ya Guo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yi-Qi Dai
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Qiu-Yan Zhao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Ming-Jie Jiang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Zhan-Jun Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| |
Collapse
|
47
|
Zhou Y, Xu Z, Lin W, Duan Y, Lu C, Liu W, Su W, Yan Y, Liu H, Liu L, Zhong M, Zhou J, Zhu H. Comprehensive Genomic Profiling of EBV-Positive Diffuse Large B-cell Lymphoma and the Expression and Clinicopathological Correlations of Some Related Genes. Front Oncol 2019; 9:683. [PMID: 31403034 PMCID: PMC6669985 DOI: 10.3389/fonc.2019.00683] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/10/2019] [Indexed: 02/05/2023] Open
Abstract
Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphoma (EBV+ DLBCL) is a rare type of lymphoma with a high incidence in elderly patients, poor drug response, and unfavorable prognosis. Despite advances in genomic profiling and precision medicine in DLBCL, EBV+ DLBCL remain poorly characterized and understood. We include 236 DLBCL patients for EBV-encoded mRNA (EBER) in situ hybridization detection and analyzed 9 EBV+ and 6 EBV negative cases by next-generation sequencing (NGS). We then performed fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) to analyze chromosome rearrangements and gene expressions in 22 EBV+ and 30 EBV negative cases. The EBER results showed a 9.3% (22/236) positive rate. The NGS results revealed recurrent alterations in MYC and RHOA, components of apoptosis and NF-κB pathways. The most frequently mutated genes in EBV+ DLBCL were MYC (3/9; 33.3%), RHOA (3/9; 33.3%), PIM1 (2/9; 22.2%), MEF2B (2/9; 22.2%), MYD88 (2/9; 22.2%), and CD79B (2/9; 22.2%) compared with KMT2D (4/6; 66.7%), CREBBP (3/6; 50.0%), PIM1 (2/6; 33.3%), TNFAIP3 (2/6; 33.3%), and BCL2 (2/6; 33.3%) in EBV-negative DLBCL. MYC and KMT2D alterations stood out the most differently mutated genes between the two groups. FISH detection displayed a lower rearrangement rate in EBV+ cohort. Furthermore, KMT2D expression was highly expressed and associated with poor survival in both cohorts. MYC was only overexpressed and related to an inferior prognosis in the EBV+ DLBCL cohort. In summary, we depicted a distinct mutation profile for EBV+ and EBV-negative DLBCL and validated the differential expression of KMT2D and MYC with potential prognostic influence, thereby providing new perspectives into the pathogenesis and precision medicine of DLBCL.
Collapse
Affiliation(s)
- Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Weiping Su
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Li Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Meizuo Zhong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
48
|
Gao LM, Zhao S, Zhang WY, Wang M, Li HF, Lizaso A, Liu WP. Somatic mutations in KMT2D and TET2 associated with worse prognosis in Epstein-Barr virus-associated T or natural killer-cell lymphoproliferative disorders. Cancer Biol Ther 2019; 20:1319-1327. [PMID: 31311407 DOI: 10.1080/15384047.2019.1638670] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Li-Min Gao
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Sha Zhao
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Wen-Yan Zhang
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Mi Wang
- Department of Dermatology and Venerology, West China Hospital of Sichuan University, Chengdu, China
| | - Hui-Fang Li
- Cellular Biology Laboratory of Core Facility, West China Hospital of Sichuan University, Chengdu, China
| | - Anle Lizaso
- Department of Medicine, Burning Rock Biotech, Shanghai, China
| | - Wei-Ping Liu
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Koutsioumpa M, Hatziapostolou M, Polytarchou C, Tolosa EJ, Almada LL, Mahurkar-Joshi S, Williams J, Tirado-Rodriguez B, Huerta-Yepez S, Karavias D, Kourea H, Poultsides GA, Struhl K, Dawson DW, Donahue TR, Fernandez-Zapico ME, lliopoulos D. Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming. Gut 2019; 68:1271-1286. [PMID: 30337373 PMCID: PMC6697184 DOI: 10.1136/gutjnl-2017-315690] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 09/06/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Despite advances in the identification of epigenetic alterations in pancreatic cancer, their biological roles in the pathobiology of this dismal neoplasm remain elusive. Here, we aimed to characterise the functional significance of histone lysine methyltransferases (KMTs) and demethylases (KDMs) in pancreatic tumourigenesis. DESIGN DNA methylation sequencing and gene expression microarrays were employed to investigate CpG methylation and expression patterns of KMTs and KDMs in pancreatic cancer tissues versus normal tissues. Gene expression was assessed in five cohorts of patients by reverse transcription quantitative-PCR. Molecular analysis and functional assays were conducted in genetically modified cell lines. Cellular metabolic rates were measured using an XF24-3 Analyzer, while quantitative evaluation of lipids was performed by liquid chromatography-mass spectrometry (LC-MS) analysis. Subcutaneous xenograft mouse models were used to evaluate pancreatic tumour growth in vivo. RESULTS We define a new antitumorous function of the histone lysine (K)-specific methyltransferase 2D (KMT2D) in pancreatic cancer. KMT2D is transcriptionally repressed in human pancreatic tumours through DNA methylation. Clinically, lower levels of this methyltransferase associate with poor prognosis and significant weight alterations. RNAi-based genetic inactivation of KMT2D promotes tumour growth and results in loss of H3K4me3 mark. In addition, KMT2D inhibition increases aerobic glycolysis and alters the lipidomic profiles of pancreatic cancer cells. Further analysis of this phenomenon identified the glucose transporter SLC2A3 as a mediator of KMT2D-induced changes in cellular, metabolic and proliferative rates. CONCLUSION Together our findings define a new tumour suppressor function of KMT2D through the regulation of glucose/fatty acid metabolism in pancreatic cancer.
Collapse
Affiliation(s)
- Marina Koutsioumpa
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| | - Maria Hatziapostolou
- Biological Sciences, University of Southampton,
Southampton, United Kingdom;,Biosciences, School of Science and Technology, Nottingham
Trent University, Nottingham, United Kingdom
| | - Christos Polytarchou
- Interdisciplinary Biomedical Research Centre, School of
Science and Technology, Nottingham Trent University, Nottingham, United
Kingdom
| | - Ezequiel J. Tolosa
- Schulze Center for Novel Therapeutics, Division of Oncology
Research, Mayo Clinic, Rochester, MN
| | - Luciana L. Almada
- Schulze Center for Novel Therapeutics, Division of Oncology
Research, Mayo Clinic, Rochester, MN
| | - Swapna Mahurkar-Joshi
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| | - Jennifer Williams
- Department of Surgery, Division of General Surgery, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | - Belen Tirado-Rodriguez
- Unidad de Investigacion en Enfermedades Oncologicas,
Hospital Infantil de Mexico, Mexico City, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigacion en Enfermedades Oncologicas,
Hospital Infantil de Mexico, Mexico City, Mexico
| | - Dimitrios Karavias
- Department of Pathology, School of Medicine, University of
Patras, Patras, Greece
| | - Helen Kourea
- Department of Pathology, School of Medicine, University of
Patras, Patras, Greece
| | | | - Kevin Struhl
- Department of Biological Chemistry and Molecular
Pharmacology, Harvard Medical School, Boston, MA
| | - David W. Dawson
- Department of Pathology and Laboratory Medicine, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | - Timothy R. Donahue
- Department of Surgery, Division of General Surgery, David
Geffen School of Medicine at University of California Los Angeles, Los Angeles,
CA
| | | | - Dimitrios lliopoulos
- Center for Systems Biomedicine, Vatche and Tamar Manoukian
Division of Digestive Diseases, David Geffen School of Medicine, University of
California at Los Angeles, Los Angeles, CA
| |
Collapse
|
50
|
Fagan RJ, Dingwall AK. COMPASS Ascending: Emerging clues regarding the roles of MLL3/KMT2C and MLL2/KMT2D proteins in cancer. Cancer Lett 2019; 458:56-65. [PMID: 31128216 DOI: 10.1016/j.canlet.2019.05.024] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022]
Abstract
The KMT2 (lysine methyltransferase) family of histone modifying proteins play essential roles in regulating developmental pathways, and mutations in the genes encoding these proteins have been strongly linked to many blood and solid tumor cancers. The KMT2A-D proteins are histone 3 lysine 4 (H3K4) methyltransferases embedded in large COMPASS-like complexes important for RNA Polymerase II-dependent transcription. KMT2 mutations were initially associated with pediatric Mixed Lineage Leukemias (MLL) and found to be the result of rearrangements of the MLL1/KMT2A gene at 11q23. Over the past several years, large-scale tumor DNA sequencing studies have revealed the potential involvement of other KMT2 family genes, including heterozygous somatic mutations in the paralogous MLL3/KMT2C and MLL2(4)/KMT2D genes that are now among the most frequently associated with human cancer. Recent studies have provided a better understanding of the potential roles of disrupted KMT2C and KMT2D family proteins in cell growth aberrancy. These findings, together with an examination of cancer genomics databases provide new insights into the contribution of KMT2C/D proteins in epigenetic gene regulation and links to carcinogenesis.
Collapse
Affiliation(s)
- Richard J Fagan
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60521, USA
| | - Andrew K Dingwall
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60521, USA; Department of Cancer Biology and Pathology & Laboratory Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, 60521, USA.
| |
Collapse
|