1
|
Nguyen HAT, Ho TP, Mangelings D, Van Eeckhaut A, Vander Heyden Y, Tran HTM. Antioxidant, neuroprotective, and neuroblastoma cells (SH-SY5Y) differentiation effects of melanins and arginine-modified melanins from Daedaleopsis tricolor and Fomes fomentarius. BMC Biotechnol 2024; 24:89. [PMID: 39529092 PMCID: PMC11556217 DOI: 10.1186/s12896-024-00918-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Microbial melanins possess a broad spectrum of biological activities. However, there is little understanding of their neuroprotective and neuronal cell differentiation properties. This study aimed to extract, purify, and modify melanins from two medicinal fungi (Daedaleopsis tricolor and Fomes fomentarius), and to evaluate their antioxidant activity, as well as their cell protective ability against neurotoxins. In addition, the study also investigated the feasibility of combining melanins or modified melanins with retinoic acid (RA) to induce neuronal differentiation. METHODS Melanin was extracted and purified using alkaline acid-based methods. Antioxidant activities and neuroprotective effects were evaluated using the DPPH (1,1-diphenyl-2-picrylhydrazyl) and MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assays, respectively. In addition, morphological changes of SH-SY5Y cells were recorded by using a Pannoramic MIDI scanner. RESULTS All melanins and arginine-modified melanins displayed mild DPPH scavenging activities, which were statistically lower than that of ascorbic acid (p < 0.05). In terms of neuroprotection, both melanins and arginine-modified melanins exhibited significant cell protection against H2O2 after 24 h exposure (p < 0.05). Notably, there is no significant difference between F. fomentarius melanin and its modified form as they both increased cell viability by about 20%. Contrarily, while D. tricolor melanin enhanced the cell viability with 16%, its modified form increased the cell viability with 21%. These activities, however, are significantly lower than the positive control (N-acetylcysteine, p < 0.05). Regarding MPTP, only the arginine-modified melanins of the two fungi significantly protected the cells after 24 h exposure to the toxin (p < 0.05). Specifically, F. fomentarius and D. tricolor modified melanins enhanced the cell viability with 10.2% and 11.1%, respectively, whereas that of the positive control was 13.2%. Interestingly, combining RA (10 µM) with 20 µg/mL of either F. fomentarius, or especially D. tricolor arginine-modified melanin, significantly promoted neuroblastoma cell differentiation into mature neuronal cells compared to using RA alone (p < 0.05). CONCLUSIONS The arginine-modified melanins of D. tricolor and F. fomentarius have potential for neuroprotection against Parkinsonian neurotoxins. In addition, the arginine-modified melanin of D. tricolor may serve as an excellent material for research in neuroblastoma treatment.
Collapse
Affiliation(s)
- Hoang Anh Thu Nguyen
- International University - VNU HCM, Applied Microbiology Laboratory, School of Biotechnology, Quarter 6, Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Thien Phu Ho
- International University - VNU HCM, Applied Microbiology Laboratory, School of Biotechnology, Quarter 6, Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Debby Mangelings
- Vrije Universiteit Brussel (VUB), Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Ann Van Eeckhaut
- Vrije Universiteit Brussel (VUB), Research Group Experimental Pharmacology (EFAR), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Center for Neurosciences (C4N), Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Yvan Vander Heyden
- Vrije Universiteit Brussel (VUB), Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Laarbeeklaan 103, Brussels, 1090, Belgium.
| | - Hanh T M Tran
- International University - VNU HCM, Applied Microbiology Laboratory, School of Biotechnology, Quarter 6, Linh Trung Ward, Thu Duc District, Ho Chi Minh City, Vietnam.
- Vrije Universiteit Brussel (VUB), Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, Laarbeeklaan 103, Brussels, 1090, Belgium.
- Vrije Universiteit Brussel (VUB), Research Group Experimental Pharmacology (EFAR), Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Center for Neurosciences (C4N), Laarbeeklaan 103, Brussels, 1090, Belgium.
| |
Collapse
|
2
|
Guha D, Singh V, Nandi S, Ramos EI, Gadad SS, Das C. ZMYND8 Is a Regulator of Sonic Hedgehog Signaling in ATRA-Mediated Differentiation of Neuroblastoma Cells. Biochemistry 2024; 63:1534-1542. [PMID: 38804064 DOI: 10.1021/acs.biochem.4c00145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Zinc Finger MYND (Myeloid, Nervy, and DEAF-1) type containing 8 (ZMYND8) is a crucial epigenetic regulator that plays a multifaceted role in governing a spectrum of vital cellular processes, encompassing proliferation, apoptosis, migration, tumor suppression, and differentiation. It has emerged as a key player in neuronal differentiation by orchestrating the expression of neuronal lineage-committed genes. The present study uncovers the role of ZMYND8 in regulating the Sonic Hedgehog (SHH) signaling axis, which is crucial for neuronal differentiation. Genetic deletion of ZMYND8 leads to a significant reduction in SHH pathway genes, GLI1, and PTCH1 expression during all-trans-retinoic acid (ATRA)-induced differentiation. ZMYND8 and RNA pol II S5P are found to co-occupy the GLI1 and PTCH1 gene promoters, positively impacting their gene transcription upon ATRA treatment. Interestingly, ZMYND8 is found to counteract the inhibitory effects of Cyclopamine that block the upstream SHH pathway protein SMO, resulting in enhanced neurite formation in neuroblastoma cells following their treatment with ATRA. These results indicate that ZMYND8 is an epigenetic regulator of the SHH signaling pathway and has tremendous therapeutic potential in ATRA-mediated differentiation of neuroblastoma.
Collapse
Affiliation(s)
- Deblina Guha
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-I, Block-AF, Bidhannagar, Kolkata 700064, West Bengal, India
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-I, Block-AF, Bidhannagar, Kolkata 700064, West Bengal, India
- Homi Bhabha National Institute, 2nd Floor, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-I, Block-AF, Bidhannagar, Kolkata 700064, West Bengal, India
- Homi Bhabha National Institute, 2nd Floor, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| | - Enrique I Ramos
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, 5001 El Paso Drive, El Paso, Texas 79905, United States
| | - Shrikanth S Gadad
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, 5001 El Paso Drive, El Paso, Texas 79905, United States
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, Texas 78229, United States
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-I, Block-AF, Bidhannagar, Kolkata 700064, West Bengal, India
- Homi Bhabha National Institute, 2nd Floor, BARC Training School Complex, Anushaktinagar, Mumbai 400094, Maharashtra, India
| |
Collapse
|
3
|
Veschi V, Durinck K, Thiele CJ, Speleman F. Neuroblastoma Epigenetic Landscape: Drugging Opportunities. PEDIATRIC ONCOLOGY 2024:71-95. [DOI: 10.1007/978-3-031-51292-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Wang L, Tang J. SWI/SNF complexes and cancers. Gene 2023; 870:147420. [PMID: 37031881 DOI: 10.1016/j.gene.2023.147420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
Epigenetics refers to the study of genetic changes that can affect gene expression without altering the underlying DNA sequence, including DNA methylation, histone modification, chromatin remodelling, X chromosome inactivation and non-coding RNA regulation. Of these, DNA methylation, histone modification and chromatin remodelling constitute the three classical modes of epigenetic regulation. These three mechanisms alter gene transcription by adjusting chromatin accessibility, thereby affecting cell and tissue phenotypes in the absence of DNA sequence changes. In the presence of ATP hydrolases, chromatin remodelling alters the structure of chromatin and thus changes the transcription level of DNA-guided RNA. To date, four types of ATP-dependent chromatin remodelling complexes have been identified in humans, namely SWI/SNF, ISWI, INO80 and NURD/MI2/CHD. SWI/SNF mutations are prevalent in a wide variety of cancerous tissues and cancer-derived cell lines as discovered by next-generation sequencing technologies.. SWI/SNF can bind to nucleosomes and use the energy of ATP to disrupt DNA and histone interactions, sliding or ejecting histones, altering nucleosome structure, and changing transcriptional and regulatory mechanisms. Furthermore, mutations in the SWI/SNF complex have been observed in approximately 20% of all cancers. Together, these findings suggest that mutations targeting the SWI/SNF complex may have a positive impact on tumorigenesis and cancer progression.
Collapse
Affiliation(s)
- Liyuan Wang
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Oncology and Hematology, Jinan 250000, Shandong Province, China
| | - Jinglong Tang
- Adicon Medical Laboratory Center, Molecular Genetic Diagnosis Center, Pathological Diagnosis Center, Jinan 250014, Shandong Province, China.
| |
Collapse
|
5
|
Chaluts D, Dullea JT, Ali M, Vasan V, Devarajan A, Rutland JW, Gill CM, Ellis E, Kinoshita Y, McBride RB, Bederson J, Donovan M, Sebra R, Umphlett M, Shrivastava RK. ARID1A mutation associated with recurrence and shorter progression-free survival in atypical meningiomas. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04442-y. [DOI: 10.1007/s00432-022-04442-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
|
6
|
Reske JJ, Wilson MR, Armistead B, Harkins S, Perez C, Hrit J, Adams M, Rothbart SB, Missmer SA, Fazleabas AT, Chandler RL. ARID1A-dependent maintenance of H3.3 is required for repressive CHD4-ZMYND8 chromatin interactions at super-enhancers. BMC Biol 2022; 20:209. [PMID: 36153585 PMCID: PMC9509632 DOI: 10.1186/s12915-022-01407-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/12/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND SWI/SNF (BAF) chromatin remodeling complexes regulate lineage-specific enhancer activity by promoting accessibility for diverse DNA-binding factors and chromatin regulators. Additionally, they are known to modulate the function of the epigenome through regulation of histone post-translational modifications and nucleosome composition, although the way SWI/SNF complexes govern the epigenome remains poorly understood. Here, we investigate the function of ARID1A, a subunit of certain mammalian SWI/SNF chromatin remodeling complexes associated with malignancies and benign diseases originating from the uterine endometrium. RESULTS Through genome-wide analysis of human endometriotic epithelial cells, we show that more than half of ARID1A binding sites are marked by the variant histone H3.3, including active regulatory elements such as super-enhancers. ARID1A knockdown leads to H3.3 depletion and gain of canonical H3.1/3.2 at ARID1A-bound active regulatory elements, and a concomitant redistribution of H3.3 toward genic elements. ARID1A interactions with the repressive chromatin remodeler CHD4 (NuRD) are associated with H3.3, and ARID1A is required for CHD4 recruitment to H3.3. ZMYND8 interacts with CHD4 to suppress a subset of ARID1A, CHD4, and ZMYND8 co-bound, H3.3+ H4K16ac+ super-enhancers near genes governing extracellular matrix, motility, adhesion, and epithelial-to-mesenchymal transition. Moreover, these gene expression alterations are observed in human endometriomas. CONCLUSIONS These studies demonstrate that ARID1A-containing BAF complexes are required for maintenance of the histone variant H3.3 at active regulatory elements, such as super-enhancers, and this function is required for the physiologically relevant activities of alternative chromatin remodelers.
Collapse
Affiliation(s)
- Jake J. Reske
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA
| | - Mike R. Wilson
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA
| | - Brooke Armistead
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA
| | - Shannon Harkins
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA
| | - Cristina Perez
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA
| | - Joel Hrit
- grid.251017.00000 0004 0406 2057Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503 USA
| | - Marie Adams
- grid.251017.00000 0004 0406 2057Genomics Core Facility, Van Andel Research Institute, Grand Rapids, MI 49503 USA
| | - Scott B. Rothbart
- grid.251017.00000 0004 0406 2057Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503 USA
| | - Stacey A. Missmer
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA ,grid.416230.20000 0004 0406 3236Department of Women’s Health, Spectrum Health System, Grand Rapids, MI 49341 USA
| | - Asgerally T. Fazleabas
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA ,grid.416230.20000 0004 0406 3236Department of Women’s Health, Spectrum Health System, Grand Rapids, MI 49341 USA
| | - Ronald L. Chandler
- grid.17088.360000 0001 2150 1785Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503 USA ,grid.251017.00000 0004 0406 2057Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503 USA ,grid.416230.20000 0004 0406 3236Department of Women’s Health, Spectrum Health System, Grand Rapids, MI 49341 USA
| |
Collapse
|
7
|
Amplification of CDK4 and MDM2: a detailed study of a high-risk neuroblastoma subgroup. Sci Rep 2022; 12:12420. [PMID: 35859155 PMCID: PMC9300649 DOI: 10.1038/s41598-022-16455-1] [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: 08/20/2021] [Accepted: 07/11/2022] [Indexed: 11/08/2022] Open
Abstract
In neuroblastoma, MYCN amplification and 11q-deletion are important, although incomplete, markers of high-risk disease. It is therefore relevant to characterize additional alterations that can function as prognostic and/or predictive markers. Using SNP-microarrays, a group of neuroblastoma patients showing amplification of one or multiple 12q loci was identified. Two loci containing CDK4 and MDM2 were commonly co-amplified, although amplification of either locus in the absence of the other was observed. Pharmacological inhibition of CDK4/6 with ribociclib or abemaciclib decreased proliferation in a broad set of neuroblastoma cell lines, including CDK4/MDM2-amplified, whereas MDM2 inhibition by Nutlin-3a was only effective in p53wild-type cells. Combined CDK4/MDM2 targeting had an additive effect in p53wild-type cell lines, while no or negative additive effect was observed in p53mutated cells. Most 12q-amplified primary tumors were of abdominal origin, including those of intrarenal origin initially suspected of being Wilms' tumor. An atypical metastatic pattern was also observed with low degree of bone marrow involvement, favoring other sites such as the lungs. Here we present detailed biological data of an aggressive neuroblastoma subgroup hallmarked by 12q amplification and atypical clinical presentation for which our in vitro studies indicate that CDK4 and/or MDM2 inhibition also could be beneficial.
Collapse
|
8
|
Truong DQ, Ho BT, Chau GC, Truong DK, Pham TTT, Nakagawara A, Bui CB. Collagen XI Alpha 1 (COL11A1) Expression in the Tumor Microenvironment Drives Neuroblastoma Dissemination. Pediatr Dev Pathol 2022; 25:91-98. [PMID: 34460335 DOI: 10.1177/10935266211039200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Neuroblastoma (NB) is among the most common cancers in children. A highly aggressive form of cancer, NB relies on cells in the microenvironment for dissemination particularly cancer associated fibroblast (CAFs). CAFs synthesise the extracellular matrix to create a scaffold for tumor growth thus enabling the carcinogenesis of NB, Collagen, an abundant scaffold protein produced by CAFs, has been implicated in the creation of an optimal tumor microenvironment, however, the expression profile of collagen within NB is not yet known. METHODS We characterised collagen expression within the tumor-stroma boundary by microarray and confirmed by qRT-PCR and immunohistochemistry. RESULTS The collagen marker, COL11A1, was also upregulated in NB CD45+ cells and SMA+ CAFs. Furthermore, SMA+ CAFs led to neuroblastoma cell invasion in an in vitro co-culture system which was subsequently attenuated by gene silencing COL11A1. Immunohistochemical staining of clinical tumor samples revealed that high COL11A1 expression in the stroma adjacent to tumour site, significantly associated with advanced cancer stages, age ≥18 months, undifferentiated tumor status, relapse and poor overall survival. CONCLUSION Collectively, these results suggest that a COL11A1 signature in the NB microenvironment could represent a novel target for therapeutic intervention.
Collapse
Affiliation(s)
| | - Ban Tran Ho
- Department of Paediatric Surgery, Faculty of Medicine, University of medicine and pharmacy at Hochiminh city, Vietnam.,Children Hospital 2, Ho Chi Minh City, Vietnam
| | - Gia-Cac Chau
- School of Medicine, Sungkyunkwan University, Suwon, Korea
| | - Dinh Khai Truong
- Department of Paediatric Surgery, Faculty of Medicine, University of medicine and pharmacy at Hochiminh city, Vietnam.,Children Hospital 2, Ho Chi Minh City, Vietnam
| | | | - Akira Nakagawara
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Chi-Bao Bui
- City Children's Hospital, Ho Chi Minh City, Vietnam.,Vietnam National University Ho Chi Minh city, Ho Chi Minh, Vietnam.,School of Medicine, Ho Chi Minh city, Vietnam
| |
Collapse
|
9
|
Mosquera Orgueira A, Cid López M, Peleteiro Raíndo A, Díaz Arias JÁ, Antelo Rodríguez B, Bao Pérez L, Alonso Vence N, Bendaña López Á, Abuin Blanco A, Melero Valentín P, Ferreiro Ferro R, Aliste Santos C, Fraga Rodríguez MF, González Pérez MS, Pérez Encinas MM, Bello López JL. Detection of Rare Germline Variants in the Genomes of Patients with B-Cell Neoplasms. Cancers (Basel) 2021; 13:cancers13061340. [PMID: 33809641 PMCID: PMC8001490 DOI: 10.3390/cancers13061340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The global importance of rare variants in tumorigenesis has been addressed by some pan-cancer analysis, revealing significant enrichments in protein-truncating variants affecting genes such as ATM, BRCA1/2, BRIP1, and MSH6. Germline variants can influence treatment response and contribute to the development of treatment-related second neoplasms, especially in childhood leukemia. We aimed to analyze the genomes of patients with B-cell lymphoproliferative disorders for the discovery of genes enriched in rare pathogenic variants. We discovered a significant enrichment for two genes in germline rare and dysfunctional variants. Additionally, we detected rare and likely pathogenic variants associated with disease prognosis and potential druggability, indicating a relevant role of these events in the variability of cancer phenotypes. Abstract There is growing evidence indicating the implication of germline variation in cancer predisposition and prognostication. Here, we describe an analysis of likely disruptive rare variants across the genomes of 726 patients with B-cell lymphoid neoplasms. We discovered a significant enrichment for two genes in rare dysfunctional variants, both of which participate in the regulation of oxidative stress pathways (CHMP6 and GSTA4). Additionally, we detected 1675 likely disrupting variants in genes associated with cancer, of which 44.75% were novel events and 7.88% were protein-truncating variants. Among these, the most frequently affected genes were ATM, BIRC6, CLTCL1A, and TSC2. Homozygous or germline double-hit variants were detected in 28 cases, and coexisting somatic events were observed in 17 patients, some of which affected key lymphoma drivers such as ATM, KMT2D, and MYC. Finally, we observed that variants in six different genes were independently associated with shorter survival in CLL. Our study results support an important role for rare germline variation in the pathogenesis and prognosis of B-cell lymphoid neoplasms.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
- Correspondence: ; Tel.: +34-981-950-191
| | - Miguel Cid López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - José Ángel Díaz Arias
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Beatriz Antelo Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Laura Bao Pérez
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Natalia Alonso Vence
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Ángeles Bendaña López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Aitor Abuin Blanco
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Paula Melero Valentín
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Roi Ferreiro Ferro
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Carlos Aliste Santos
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Máximo Francisco Fraga Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
- Department of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Marta Sonia González Pérez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Manuel Mateo Pérez Encinas
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - José Luis Bello López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Department of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
10
|
SWI/SNF chromatin remodeling complex alterations in meningioma. J Cancer Res Clin Oncol 2021; 147:3431-3440. [PMID: 33715086 DOI: 10.1007/s00432-021-03586-7] [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: 02/15/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE While SWI/SNF chromatin remodeling complex alterations occur in approximately 20% of cancer, the frequency and potential impact on clinical outcomes in meningiomas remains to be comprehensively elucidated. METHODS A large series of 255 meningiomas from a single institution that was enriched for high grade and recurrent lesions was identified. We performed next-generation targeted sequencing of known meningioma driver genes, including NF2, AKT1, PIK3CA, PIK3R1, and SMO and SWI/SNF chromatin remodeling complex genes, including ARID1A, SMARCA4, and SMARCB1 in all samples. Clinical correlates focused on clinical presentation and patient outcomes are presented. RESULTS The series included 63 grade I meningiomas and 192 high-grade meningiomas, including 173 WHO grade II and 19 WHO grade III. Samples from recurrent surgeries comprised 37.3% of the series. A total of 41.6% meningiomas were from the skull base. NF2, AKT1, PIK3CA, PIK3R1, and SMO were mutated in 40.8, 7.1, 3.5, 3.9, and 2.4% of samples, respectively. ARID1A, SMARCA4, and SMARCB1 mutations were observed in 17.3, 3.5, and 5.1% of samples, respectively. A total of 68.2% of ARID1A-mutant meningiomas harbored a p.Gln1327del in-frame deletion. ARID1A mutations were seen in 19.1% of Grade I, 16.8% of Grade II, and 15.8% of Grade III meningiomas (P = 0.9, Fisher's exact). Median overall survival was 16.3 years (95% CI 10.9, 16.8). With multivariable analysis, the presence of an ARID1A mutation was significantly associated with a 7.421-fold increased hazard of death (P = 0.04). CONCLUSION ARID1A mutations occur with similar frequency between low and high-grade meningiomas, but ARID1A mutations are independently prognostic of worse prognosis beyond clinical and histopathologic features.
Collapse
|
11
|
Abstract
Neuroblastoma (NB) is a pediatric cancer of the sympathetic nervous system and one of the most common solid tumors in infancy. Amplification of MYCN, copy number alterations, numerical and segmental chromosomal aberrations, mutations, and rearrangements on a handful of genes, such as ALK, ATRX, TP53, RAS/MAPK pathway genes, and TERT, are attributed as underlying causes that give rise to NB. However, the heterogeneous nature of the disease-along with the relative paucity of recurrent somatic mutations-reinforces the need to understand the interplay of genetic factors and epigenetic alterations in the context of NB. Epigenetic mechanisms tightly control gene expression, embryogenesis, imprinting, chromosomal stability, and tumorigenesis, thereby playing a pivotal role in physio- and pathological settings. The main epigenetic alterations include aberrant DNA methylation, disrupted patterns of posttranslational histone modifications, alterations in chromatin composition and/or architecture, and aberrant expression of non-coding RNAs. DNA methylation and demethylation are mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, respectively, while histone modifications are coordinated by histone acetyltransferases and deacetylases (HATs, HDACs), and histone methyltransferases and demethylases (HMTs, HDMs). This article focuses predominately on the crosstalk between the epigenome and NB, and the implications it has on disease diagnosis and treatment.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| |
Collapse
|
12
|
A C21-steroidal derivative suppresses T-cell lymphoma in mice by inhibiting SIRT3 via SAP18-SIN3. Commun Biol 2020; 3:732. [PMID: 33273692 PMCID: PMC7713351 DOI: 10.1038/s42003-020-01458-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/02/2020] [Indexed: 01/23/2023] Open
Abstract
The SIN3 repressor complex and the NAD-dependent deacetylase SIRT3 control cell growth, and development as well as malignant transformation. Even then, a little known about cross-talks between these two chromatin modifiers or whether their interaction explored therapeutically. Here we describe the identification of a C21-steroidal derivative compound, 3-O-chloroacetyl-gagamine, A671, which potently suppresses the growth of mouse and human T-cell lymphoma and erythroleukemia in vitro and preclinical models. A671 exerts its anti-neoplastic effects by direct interaction with Histone deacetylase complex subunit SAP18, a component of the SIN3 suppressor complex. This interaction stabilizes and activates SAP18, leading to transcriptional suppression of SIRT3, consequently to inhibition of proliferation and cell death. The resistance of cancer cells to A671 correlated with diminished SAP18 activation and sustained SIRT3 expression. These results uncover the SAP18-SIN3-SIRT3 axis that can be pharmacologically targeted by a C21-steroidal agent to suppress T-cell lymphoma and other malignancies. Gajendran et al. show that a C21-steroidal derivative called A671, 3-O-chloroacetyl-gagamine, suppresses the growth of T-cell lymphoma in mice. They find that A671 activates SAP18 to suppress the transcription of SIRT3, inhibiting cell growth. This study presents a new pharmacological target pathway for T-cell lymphoma.
Collapse
|