151
|
Fahey JM, Korytowski W, Girotti AW. Upstream signaling events leading to elevated production of pro-survival nitric oxide in photodynamically-challenged glioblastoma cells. Free Radic Biol Med 2019; 137:37-45. [PMID: 30991141 PMCID: PMC6526063 DOI: 10.1016/j.freeradbiomed.2019.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/15/2019] [Accepted: 04/10/2019] [Indexed: 12/19/2022]
Abstract
Nitric oxide (NO) generated endogenously by inducible nitric oxide synthase (iNOS) promotes growth and migration/invasion of glioblastoma cells and also fosters resistance to chemotherapy and ionizing radiotherapy. Our recent studies revealed that glioblastoma cell iNOS/NO also opposes the cytotoxic effects of non-ionizing photodynamic therapy (PDT), and moreover stimulates growth/migration aggressiveness of surviving cells. These negative responses, which depended on PI3K/Akt/NF-κB activation, were strongly suppressed by blocking iNOS transcription with JQ1, a BET bromodomain inhibitor. In the present study, we sought to identify additional molecular events that precede iNOS transcriptional upregulation. Akt activation, iNOS induction, and viability loss in PDT-challenged glioblastoma U87 cells were all strongly inhibited by added l-histidine, consistent with primary involvement of photogenerated singlet oxygen (1O2). Transacetylase p300 not only underwent greater Akt-dependent activation after PDT, but greater interaction with NF-κB subunit p65, which in turn exhibited greater K310 acetylation. In addition, PDT promoted intramolecular disulfide formation and inactivation of tumor suppressor PTEN, thereby favoring Akt and p300 activation leading to iNOS upregulation. Importantly, deacetylase Sirt1 was down-regulated by PDT stress, consistent with the observed increase in p65-acK310 level, which fostered iNOS transcription. This study provides new mechanistic insights into how glioblastoma tumors can exploit iNOS/NO to not only resist PDT, but to attain a more aggressive survival phenotype.
Collapse
Affiliation(s)
- Jonathan M Fahey
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226-3548, USA
| | | | - Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226-3548, USA.
| |
Collapse
|
152
|
Cinnamon and its Metabolite Protect the Nigrostriatum in a Mouse Model of Parkinson's Disease Via Astrocytic GDNF. J Neuroimmune Pharmacol 2019; 14:503-518. [PMID: 31119595 DOI: 10.1007/s11481-019-09855-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/22/2019] [Indexed: 12/11/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has potent neurotrophic effects and is known to promote the dopaminergic (DA) neuronal survival in cellular and animal models of Parkinson's disease (PD). However, long-term ectopic GDNF delivery is associated with long lasting adverse side effects in PD patients. Therefore, finding safer and effective ways to elevate endogenous GDNF levels is an active area of research. This study underlines the importance of sodium benzoate (NaB), a metabolite of commonly-used spice cinnamon, a food-additive and an FDA-approved drug against hyperammonemia, in stimulating GDNF in primary mouse and human astrocytes. Presence of cAMP response element (CRE) in the Gdnf gene promoter, recruitment of CREB to the Gdnf promoter by NaB and abrogation of NaB-mediated GDNF expression by siRNA knockdown of CREB suggest that NaB induces the transcription of Gdnf via CREB. Finally, oral administration of NaB and cinnamon itself increased the level of GDNF in vivo in the substantia nigra pars compacta (SNpc) of normal as well as MPTP-intoxicated mice. Accordingly, cinnamon and NaB treatment protected tyrosine hydroxylase positive neurons in the SNpc and fibers in the striatum, normalized striatal neurotransmitters, and improved locomotor activities in MPTP-intoxicated Gfapcre mice, but not GdnfΔastro mice lacking GDNF in astrocytes. These findings highlight the importance of astroglial GDNF in cinnamon- and NaB-mediated protection of the nigrostriatum in MPTP mouse model of PD and suggest possible therapeutic potential of cinnamon and NaB in PD patients. Graphical abstract Cinnamon metabolite sodium benzoate (NaB) activates cAMP-response element-binding (CREB) via protein kinase A (PKA) in astrocytes. Activated CREB then binds to cAMP-response element (CRE) present in GDNF gene promoter to stimulate the transcription of GDNF in astrocytes. This astrocytic GDNF leads to nigral trophism and protects dopaminergic neurons from MPTP insult.
Collapse
|
153
|
H3K18Ac as a Marker of Cancer Progression and Potential Target of Anti-Cancer Therapy. Cells 2019; 8:cells8050485. [PMID: 31121824 PMCID: PMC6562857 DOI: 10.3390/cells8050485] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
Acetylation and deacetylation are posttranslational modifications (PTMs) which affect the regulation of chromatin structure and its remodeling. Acetylation of histone 3 at lysine placed on position 18 (H3K18Ac) plays an important role in driving progression of many types of cancer, including breast, colon, lung, hepatocellular, pancreatic, prostate, and thyroid cancer. The aim of this review is to analyze and discuss the newest findings regarding the role of H3K18Ac and acetylation of other histones in carcinogenesis. We summarize the level of H3K18Ac in different cancer cell lines and analyze its association with patients’ outcomes, including overall survival (OS), progression-free survival (PFS), and disease-free survival (DFS). Finally, we describe future perspectives of cancer therapeutic strategies based on H3K18 modifications.
Collapse
|
154
|
Mata-Rocha M, Rangel-López A, Jiménez-Hernández E, Morales-Castillo BA, González-Torres C, Gaytan-Cervantes J, Álvarez-Olmos E, Núñez-Enríquez JC, Fajardo-Gutiérrez A, Martín-Trejo JA, Solís-Labastida KA, Medina-Sansón A, Flores-Lujano J, Sepúlveda-Robles OA, Peñaloza-González JG, Espinoza-Hernández LE, Núñez-Villegas NN, Espinosa-Elizondo RM, Cortés-Herrera B, Torres-Nava JR, Flores-Villegas LV, Merino-Pasaye LE, Bekker-Méndez VC, Velázquez-Aviña MM, Pérez-Saldívar ML, Bautista-Martínez BA, Amador-Sánchez R, González-Avila AI, Jiménez-Morales S, Duarte-Rodríguez DA, Santillán-Juárez JD, García-Velázquez AJ, Rosas-Vargas H, Mejía-Aranguré JM. Identification and Characterization of Novel Fusion Genes with Potential Clinical Applications in Mexican Children with Acute Lymphoblastic Leukemia. Int J Mol Sci 2019; 20:E2394. [PMID: 31096545 PMCID: PMC6566803 DOI: 10.3390/ijms20102394] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 11/16/2022] Open
Abstract
Acute lymphoblastic leukemia is the most common type of childhood cancer worldwide. Mexico City has one of the highest incidences and mortality rates of this cancer. It has previously been recognized that chromosomal translocations are important in cancer etiology. Specific fusion genes have been considered as important treatment targets in childhood acute lymphoblastic leukemia (ALL). The present research aimed at the identification and characterization of novel fusion genes with potential clinical implications in Mexican children with acute lymphoblastic leukemia. The RNA-sequencing approach was used. Four fusion genes not previously reported were identified: CREBBP-SRGAP2B, DNAH14-IKZF1, ETV6-SNUPN, ETV6-NUFIP1. Although a fusion gene is not sufficient to cause leukemia, it could be involved in the pathogenesis of the disease. Notably, these new translocations were found in genes encoding for hematopoietic transcription factors which are known to play an important role in leukemogenesis and disease prognosis such as IKZF1, CREBBP, and ETV6. In addition, they may have an impact on the prognosis of Mexican pediatric patients with ALL, with the potential to be included in the current risk stratification schemes or used as therapeutic targets.
Collapse
Affiliation(s)
- Minerva Mata-Rocha
- CONACyT-Unidad de Investigacion Medica en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Siglo XXI, IMSS, 06720 Mexico City, Mexico.
- Unidad de Investigacion Medica en Genética Humana, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Angelica Rangel-López
- Coordinacion de Investigacion en Salud, Unidad Habilitada de Apoyo al Predictamen, Centro Medico Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Elva Jiménez-Hernández
- Servicio de Hematologia Pediatrica, Hospital General "Gaudencio González Garza", Centro Medico Nacional (CMN) "La Raza", IMSS, 02990 Mexico City, Mexico.
- Servicio de Oncología, Hospital Pediatrico de Moctezuma, Secretaria de Salud de la Ciudad de Mexico, Ciudad de Mexico, 15530 Mexico City, Mexico.
| | - Blanca Angélica Morales-Castillo
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Carolina González-Torres
- Laboratorio de Secuenciación, Division de Desarrollo de la Investigacion, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Javier Gaytan-Cervantes
- Laboratorio de Secuenciación, Division de Desarrollo de la Investigacion, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Enrique Álvarez-Olmos
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Juan Carlos Núñez-Enríquez
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Arturo Fajardo-Gutiérrez
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Jorge Alfonso Martín-Trejo
- Servicio de Hematologia, UMAE Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | | | - Aurora Medina-Sansón
- Servicio de Oncología, Hospital Infantil de Mexico Federico Gómez, Secretaria de Salud, 06720 Mexico City, Mexico.
| | - Janet Flores-Lujano
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Omar Alejandro Sepúlveda-Robles
- CONACyT-Unidad de Investigacion Medica en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Siglo XXI, IMSS, 06720 Mexico City, Mexico.
- Unidad de Investigacion Medica en Genética Humana, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | | | - Laura Eugenia Espinoza-Hernández
- Servicio de Hematologia Pediatrica, Hospital General "Gaudencio González Garza", Centro Medico Nacional (CMN) "La Raza", IMSS, 02990 Mexico City, Mexico.
| | - Nora Nancy Núñez-Villegas
- Servicio de Hematologia Pediatrica, Hospital General "Gaudencio González Garza", Centro Medico Nacional (CMN) "La Raza", IMSS, 02990 Mexico City, Mexico.
| | | | - Beatriz Cortés-Herrera
- Servicio de Hematologia Pediatrica, Hospital General de Mexico, Secretaria de Salud, 06726 Mexico City, Mexico.
| | - José Refugio Torres-Nava
- Servicio de Oncología, Hospital Pediatrico de Moctezuma, Secretaria de Salud de la Ciudad de Mexico, Ciudad de Mexico, 15530 Mexico City, Mexico.
| | - Luz Victoria Flores-Villegas
- Servicio de Hematologia Pediatrica, Centro Medico Nacional "20 de Noviembre", ISSSTE, 03229 Mexico City, Mexico.
| | | | - Vilma Carolina Bekker-Méndez
- Unidad de Investigacion Medico en Inmunologia e Infectologia, Hospital de Infectologia "Dr. Daniel Méndez Hernández", "La Raza", IMSS, 02990 Mexico City, Mexico.
| | | | - María Luisa Pérez-Saldívar
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | | | - Raquel Amador-Sánchez
- Servicio de Hematologia Pediatrica, Hospital General Regional "Carlos McGregor Sanchez Navarro", IMSS, 03100 Mexico City, Mexico.
| | - Ana Itamar González-Avila
- Servicio de Hematologia Pediatrica, Hospital General Regional "Carlos McGregor Sanchez Navarro", IMSS, 03100 Mexico City, Mexico.
| | - Silvia Jiménez-Morales
- Laboratorio de Genomica del Cancer del Instituto Nacional de Medicina Genomica (INMEGEN), 14610 Mexico City, Mexico.
| | - David Aldebarán Duarte-Rodríguez
- Unidad de Investigacion Medico en Epidemiologia Clinica, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | | | | | - Haydeé Rosas-Vargas
- Unidad de Investigacion Medica en Genética Humana, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, IMSS, 06720 Mexico City, Mexico.
| | - Juan Manuel Mejía-Aranguré
- Coordinación de Investigacion en Salud, IMSS, Torre Academia Nacional de Medicina, 06720 Mexico City, Mexico.
| |
Collapse
|
155
|
Hu Y, Zhang Q, Hu D, Wang J, Rao J, Xu L, Guo Z, Wang S, Liu X, Tang S, Shen Q. Ultrasensitive electrochemiluminescence immunosensor for the transcriptional co-activator p300 by using a graphene oxide monolayer and tetrahedral DNA-mediated signal amplification. Mikrochim Acta 2019; 186:325. [DOI: 10.1007/s00604-019-3435-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/11/2019] [Indexed: 01/21/2023]
|
156
|
Liu H, Liu H, Li X. Use of Serine/Threonine Ligation for the Total Chemical Synthesis of HMGA1a Protein with Site‐Specific Lysine Acetylations. Chempluschem 2019; 84:779-785. [DOI: 10.1002/cplu.201900130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/10/2019] [Indexed: 01/21/2023]
Affiliation(s)
- Heng Liu
- Department of Chemistry State Key Laboratory of Synthetic ChemistryThe University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| | - Han Liu
- Department of Chemistry State Key Laboratory of Synthetic ChemistryThe University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| | - Xuechen Li
- Department of Chemistry State Key Laboratory of Synthetic ChemistryThe University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| |
Collapse
|
157
|
Padmanabhan A, Haldar SM. Drugging transcription in heart failure. J Physiol 2019; 598:3005-3014. [PMID: 30927446 DOI: 10.1113/jp276745] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Advances in our understanding of the basic biology and biochemistry of chromatin structure and function at genome scales has led to tremendous growth in the fields of epigenomics and transcriptional biology. While it has long been appreciated that transcriptional pathways are dysregulated in failing hearts, only recently has the idea of disrupting altered transcription by targeting chromatin-associated proteins been explored. Here, we provide a brief overview of efforts to drug transcription in the context of heart failure, focusing on the bromo- and extra-terminal domain (BET) family of chromatin co-activator proteins.
Collapse
Affiliation(s)
- Arun Padmanabhan
- Division of Cardiology, Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA.,Gladstone Institutes, San Francisco, CA, USA
| | - Saptarsi M Haldar
- Division of Cardiology, Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA.,Gladstone Institutes, San Francisco, CA, USA.,Cardiometabolic Disorders, Amgen, South San Francisco, CA, USA
| |
Collapse
|
158
|
Bi X, Xu M, Li J, Huang T, Jiang B, Shen L, Luo L, Liu S, Yin Z. Heat shock protein 27 inhibits HMGB1 translocation by regulating CBP acetyltransferase activity and ubiquitination. Mol Immunol 2019; 108:45-55. [DOI: 10.1016/j.molimm.2019.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 11/28/2022]
|
159
|
Hernández-Acevedo GN, López-Portales OH, Gutiérrez-Reyna DY, Cuevas-Fernández E, Kempis-Calanis LA, Labastida-Conde RG, Aguilar-Luviano OB, Ramírez-Pliego O, Spicuglia S, Lino-Alfaro B, Chagolla-López A, González-de la Vara LE, Santana MA. Protein complexes associated with β-catenin differentially influence the differentiation profile of neonatal and adult CD8 + T cells. J Cell Physiol 2019; 234:18639-18652. [PMID: 30924167 DOI: 10.1002/jcp.28502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 02/13/2019] [Accepted: 02/19/2019] [Indexed: 02/04/2023]
Abstract
The canonical Wnt signaling pathway is a master cell regulator involved in CD8+ T cell proliferation and differentiation. In human CD8+ T cells, this pathway induces differentiation into memory cells or a "stem cell memory like" population, which is preferentially present in cord blood. To better understand the role of canonical Wnt signals in neonatal or adult blood, we compared the proteins associated with β-catenin, in nonstimulated and Wnt3a-stimulated human neonatal and adult naive CD8+ T cells. Differentially recruited proteins established different complexes in adult and neonatal cells. In the former, β-catenin-associated proteins were linked to cell signaling and immunological functions, whereas those of neonates were linked to proliferation and metabolism. Wnt3a stimulation led to the recruitment and overexpression of Wnt11 in adult cells and Wnt5a in neonatal cells, suggesting a differential connexion with planar polarity and Wnt/Ca2+ noncanonical pathways, respectively. The chromatin immunoprecipitation polymerase chain reaction β-catenin was recruited to a higher level on the promoters of cell renewal genes in neonatal cells and of differentiation genes in those of adults. We found a preferential association of β-catenin with CBP in neonatal cells and with p300 in the adult samples, which could be involved in a higher self-renewal capacity of the neonatal cells and memory commitment in those of adults. Altogether, our results show that different proteins associated with β-catenin during Wnt3a activation mediate a differential response of neonatal and adult human CD8+ T cells.
Collapse
Affiliation(s)
- Gerson N Hernández-Acevedo
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Oscar H López-Portales
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Darely Y Gutiérrez-Reyna
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Erick Cuevas-Fernández
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Linda A Kempis-Calanis
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Rosario G Labastida-Conde
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Oscar B Aguilar-Luviano
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Oscar Ramírez-Pliego
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Salvatore Spicuglia
- Aix-Marseille University, Inserm, TAGC, UMR1090, Marseille, France.,Equipe Labélisée Ligue Contre le Cancer, Marseille, France
| | - Bárbara Lino-Alfaro
- Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Irapuato, Mexico
| | - Alicia Chagolla-López
- Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Irapuato, Mexico
| | | | - María Angélica Santana
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| |
Collapse
|
160
|
Biswas S, Chakrabarti S. Increased Extracellular Matrix Protein Production in Chronic Diabetic Complications: Implications of Non-Coding RNAs. Noncoding RNA 2019; 5:E30. [PMID: 30909482 PMCID: PMC6468528 DOI: 10.3390/ncrna5010030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Management of chronic diabetic complications remains a major medical challenge worldwide. One of the characteristic features of all chronic diabetic complications is augmented production of extracellular matrix (ECM) proteins. Such ECM proteins are deposited in all tissues affected by chronic complications, ultimately causing organ damage and dysfunction. A contributing factor to this pathogenetic process is glucose-induced endothelial damage, which involves phenotypic transformation of endothelial cells (ECs). This phenotypic transition of ECs, from a quiescent state to an activated dysfunctional state, can be mediated through alterations in the synthesis of cellular proteins. In this review, we discussed the roles of non-coding RNAs, specifically microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in such processes. We further outlined other epigenetic mechanisms regulating the biogenesis and/or function of non-coding RNAs. Overall, we believe that better understanding of such molecular processes may lead to the development of novel biomarkers and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Saumik Biswas
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| |
Collapse
|
161
|
Lee J, Lee TH. How Protein Binding Sensitizes the Nucleosome to Histone H3K56 Acetylation. ACS Chem Biol 2019; 14:506-515. [PMID: 30768236 DOI: 10.1021/acschembio.9b00018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The nucleosome, the fundamental gene-packing unit comprising an octameric histone protein core wrapped with DNA, has a flexible structure that enables dynamic gene regulation mechanisms. Histone lysine acetylation at H3K56 removes a positive charge from the histone core where it interacts with the termini of the nucleosomal DNA and acts as a critical gene regulatory signal that is implicated in transcription initiation and elongation. The predominant proposal for the biophysical role of H3K56 acetylation (H3K56ac) is that weakened electrostatic interaction between DNA termini and the histone core results in facilitated opening and subsequent disassembly of the nucleosome. However, this effect alone is too weak to account for the strong coupling between H3K56ac and its regulatory outcomes. Here we utilized a semisynthetically modified nucleosome with H3K56ac in order to address this discrepancy. Based on the results, we propose an innovative mechanism by which the charge neutralization effect of H3K56ac is significantly amplified via protein binding. We employed three-color single-molecule fluorescence resonance energy transfer (smFRET) to monitor the opening rate of nucleosomal DNA termini induced by binding of histone chaperone Nap1. We observed an elevated opening rate upon H3K56ac by 5.9-fold, which is far larger than the 1.5-fold previously reported for the spontaneous opening dynamics in the absence of Nap1. Our proposed mechanism successfully reconciles this discrepancy because DNA opening for Nap1 binding must be larger than the average spontaneous opening. This is a novel mechanism that can explain how a small biophysical effect of histone acetylation results in a significant change in protein binding rate.
Collapse
Affiliation(s)
- Jaehyoun Lee
- Department of Chemistry, the Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Tae-Hee Lee
- Department of Chemistry, the Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
162
|
CBP and SRF co-regulate dendritic growth and synaptic maturation. Cell Death Differ 2019; 26:2208-2222. [PMID: 30850733 PMCID: PMC6889142 DOI: 10.1038/s41418-019-0285-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/17/2018] [Accepted: 01/10/2019] [Indexed: 12/03/2022] Open
Abstract
The CREB-binding protein (CBP) exerts tight control of developmental processes. Here, we investigated the consequences of its selective ablation in newborn neurons. Mice in which CBP was eliminated during neuronal differentiation showed perinatal death and defective diaphragm innervation. Adult-born neurons also showed impaired growth and maturation after inducible and restricted CBP loss in dentate gyrus neuroprogenitors. Consistent with these in vivo findings, cultured neurons displayed impaired outgrowth, immature spines, and deficient activity-dependent synaptic remodeling after CBP ablation. These deficits coincided with broad transcriptional changes affecting genes involved in neuronal growth and plasticity. The affected gene set included many predicted targets of both CBP and the serum response factor (SRF), an activity-regulated transcription factor involved in structural plasticity. Notably, increasing SRF activity in a CBP-independent manner ameliorated the transcriptional, synaptic, and growth defects. These results underscore the relevance of CBP–SRF interactions during neuronal outgrowth and synaptic maturation, and demonstrate that CBP plays an essential role in supporting the gene program underlying the last steps of neuronal differentiation, both during development and in the adult brain.
Collapse
|
163
|
Berlow RB, Martinez-Yamout MA, Dyson HJ, Wright PE. Role of Backbone Dynamics in Modulating the Interactions of Disordered Ligands with the TAZ1 Domain of the CREB-Binding Protein. Biochemistry 2019; 58:1354-1362. [PMID: 30775911 DOI: 10.1021/acs.biochem.8b01290] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The intrinsically disordered transactivation domains of HIF-1α and CITED2 compete for binding of the TAZ1 domain of the CREB-binding protein by a unidirectional allosteric mechanism involving direct competition for shared binding sites, ternary complex formation, and TAZ1 conformational changes. To gain insight into the mechanism by which CITED2 displaces HIF-1α from TAZ1, we used nuclear magnetic resonance spin relaxation methods to obtain an atomic-level description of the picosecond to nanosecond backbone dynamics that contribute to TAZ1 binding and competition. We show that HIF-1α and CITED2 adopt different dynamics in their complexes with TAZ1, with flexibility observed for HIF-1α in regions that would maintain accessibility for CITED2 to bind to TAZ1 and facilitate subsequent HIF-1α dissociation. In contrast, critical regions of CITED2 adopt a rigid structure in its complex with TAZ1, minimizing the ability of HIF-1α to compete for binding. We also find that TAZ1, previously thought to be a rigid scaffold for binding of disordered protein ligands, displays altered backbone dynamics in its various bound states. TAZ1 is more rigid in its CITED2-bound state than in its free state or in complex with HIF-1α, with increased rigidity observed not only in the CITED2 binding site but also in regions of TAZ1 that undergo conformational changes between the HIF-1α- and CITED2-bound structures. Taken together, these data suggest that backbone dynamics in TAZ1, as well as in the HIF-1α and CITED2 ligands, play a role in modulating the occupancy of TAZ1 and highlight the importance of characterizing both binding partners in molecular interactions.
Collapse
Affiliation(s)
- Rebecca B Berlow
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Maria A Martinez-Yamout
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - H Jane Dyson
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Peter E Wright
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| |
Collapse
|
164
|
Abstract
IMPACT STATEMENT This review provides various genetic and cell line data previously published in a way to explain how cellular stress can lead into genetic instability.
Collapse
Affiliation(s)
- Jung Joo Moon
- 1 JS Yoon Memorial Cancer Research Institute LLC, Lutherville, MD 2109, USA
| | - Alexander Lu
- 1 JS Yoon Memorial Cancer Research Institute LLC, Lutherville, MD 2109, USA
| | - Chulso Moon
- 1 JS Yoon Memorial Cancer Research Institute LLC, Lutherville, MD 2109, USA.,2 Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institution, Baltimore, MD 21205, USA
| |
Collapse
|
165
|
Zhu YX, Shi CX, Bruins LA, Wang X, Riggs DL, Porter B, Ahmann JM, de Campos CB, Braggio E, Bergsagel PL, Stewart AK. Identification of lenalidomide resistance pathways in myeloma and targeted resensitization using cereblon replacement, inhibition of STAT3 or targeting of IRF4. Blood Cancer J 2019; 9:19. [PMID: 30741931 PMCID: PMC6370766 DOI: 10.1038/s41408-019-0173-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 11/29/2022] Open
Abstract
To understand immunomodulatory drug (IMiD) resistance in multiple myeloma (MM), we created isogenic human multiple myeloma cell lines (HMCLs) sensitive and resistant to lenalidomide, respectively. Four HMCLs were demonstrated to be resistant to all IMiDs including lenalidomide, pomalidomide, and CC-220, but not to Bortezomib. In three HMLCs (MM.1.SLenRes, KMS11LenRes and OPM2LenRes), CRBN abnormalities were found, including chromosomal deletion, point mutation, and low CRBN expression. The remaining HMCL, XG1LenRes, showed no changes in CRBN but exhibited CD147 upregulation and impaired IRF4 downregulation after lenalidomide treatment. Depletion of CD147 in XG1LenRes and three additional HMCLs had no significant impact on MM viability and lenalidomide response. Further analysis of XG1LenRes demonstrated increased IL6 expression and constitutive STAT3 activation. Inhibition of STAT3 with a selective compound (PB-1-102) re-sensitized XG1LenRes to lenalidomide. Since XG1LenRes harbors a truncated IRF4 that is not downregulated by lenalidomide, we targeted IRF4/MYC axis with a selective inhibitor of the bromodomain of CBP/EP300 (SGC-CBP30), which restored lenalidomide response in XG1LenRes. This strategy also appeared to be more broadly applicable as SGC-CBP30 could re-sensitize two resistant HMCLs with low but detectable CRBN expression to lenalidomide, suggesting that targeting CBP/E300 is a promising approach to restore IMiD sensitivity in MM with detectable CRBN expression.
Collapse
Affiliation(s)
- Yuan Xiao Zhu
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Chang-Xin Shi
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Laura A Bruins
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Xuewei Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Daniel L Riggs
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | - Brooke Porter
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA
| | | | | | | | | | - A Keith Stewart
- Division of Hematology, Mayo Clinic, Scottsdale, AZ, USA. .,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
166
|
Angius A, Uva P, Oppo M, Persico I, Onano S, Olla S, Pes V, Perria C, Cuccuru G, Atzeni R, Serra G, Cucca F, Sotgiu S, Hennekam RC, Crisponi L. Confirmation of a new phenotype in an individual with a variant in the last part of exon 30 of CREBBP. Am J Med Genet A 2019; 179:634-638. [PMID: 30737887 DOI: 10.1002/ajmg.a.61052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/28/2018] [Accepted: 01/01/2019] [Indexed: 11/10/2022]
Abstract
We report here a novel de novo missense variant affecting the last amino acid of exon 30 of CREBBP [NM_004380, c.5170G>A; p.(Glu1724Lys)] in a 17-year-old boy presenting mild intellectual disability and dysmorphisms but not resembling the phenotype of classical Rubinstein-Taybi syndrome. The patient showed a marked overweight from early infancy on and had cortical heterotopias. Recently, 22 individuals have been reported with missense mutations in the last part of exon 30 and the beginning of exon 31 of CREBBP, showing this new phenotype. This additional case further delineates the genotype-phenotype correlations within the molecular and phenotypic spectrum of variants in CREBBP and EP300.
Collapse
Affiliation(s)
- Andrea Angius
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy
| | - Paolo Uva
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula (CA), Italy
| | - Manuela Oppo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Ivana Persico
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy
| | - Stefano Onano
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Stefania Olla
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy
| | - Valentina Pes
- Clinica di Neuropsichiatria Infantile, Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy
| | - Chiara Perria
- Clinica di Neuropsichiatria Infantile, Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy
| | - Gianmauro Cuccuru
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula (CA), Italy
| | - Rossano Atzeni
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Science and Technology Park Polaris, Pula (CA), Italy
| | - Gigliola Serra
- Clinica di Neuropsichiatria Infantile, Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Stefano Sotgiu
- Clinica di Neuropsichiatria Infantile, Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Sassari, Italy
| | - Raoul C Hennekam
- Department of Pediatrics, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Laura Crisponi
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato (CA), Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| |
Collapse
|
167
|
Leatham-Jensen M, Uyehara CM, Strahl BD, Matera AG, Duronio RJ, McKay DJ. Lysine 27 of replication-independent histone H3.3 is required for Polycomb target gene silencing but not for gene activation. PLoS Genet 2019; 15:e1007932. [PMID: 30699116 PMCID: PMC6370247 DOI: 10.1371/journal.pgen.1007932] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/11/2019] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Proper determination of cell fates depends on epigenetic information that is used to preserve memory of decisions made earlier in development. Post-translational modification of histone residues is thought to be a central means by which epigenetic information is propagated. In particular, modifications of histone H3 lysine 27 (H3K27) are strongly correlated with both gene activation and gene repression. H3K27 acetylation is found at sites of active transcription, whereas H3K27 methylation is found at loci silenced by Polycomb group proteins. The histones bearing these modifications are encoded by the replication-dependent H3 genes as well as the replication-independent H3.3 genes. Owing to differential rates of nucleosome turnover, H3K27 acetylation is enriched on replication-independent H3.3 histones at active gene loci, and H3K27 methylation is enriched on replication-dependent H3 histones across silenced gene loci. Previously, we found that modification of replication-dependent H3K27 is required for Polycomb target gene silencing, but it is not required for gene activation. However, the contribution of replication-independent H3.3K27 to these functions is unknown. Here, we used CRISPR/Cas9 to mutate the endogenous replication-independent H3.3K27 to a non-modifiable residue. Surprisingly, we find that H3.3K27 is also required for Polycomb target gene silencing despite the association of H3.3 with active transcription. However, the requirement for H3.3K27 comes at a later stage of development than that found for replication-dependent H3K27, suggesting a greater reliance on replication-independent H3.3K27 in post-mitotic cells. Notably, we find no evidence of global transcriptional defects in H3.3K27 mutants, despite the strong correlation between H3.3K27 acetylation and active transcription. During development, naïve precursor cells acquire distinct identities through differential regulation of gene expression. The process of cell fate specification is progressive and depends on memory of prior developmental decisions. Maintaining cell identities over time is not dependent on changes in genome sequence. Instead, epigenetic mechanisms propagate information on cell identity by maintaining select sets of genes in either the on or off state. Chemical modifications of histone proteins, which package and organize the genome within cells, are thought to play a central role in epigenetic gene regulation. However, identifying which histone modifications are required for gene regulation, and defining the mechanisms through which they function in the maintenance of cell identity, remains a longstanding research challenge. Here, we focus on the role of histone H3 lysine 27 (H3K27). Modifications of H3K27 are associated with both gene activation and gene silencing (i.e. H3K27 acetylation and methylation, respectively). The histones bearing these modifications are encoded by different histone genes. One set of histone genes is only expressed during cell division, whereas the other set of histone genes is expressed in both dividing and non-dividing cells. Because most cells permanently stop dividing by the end of development, these “replication-independent” histone genes are potentially important for long-term maintenance of cell identity. In this study, we demonstrate that replication-independent H3K27 is required for gene silencing by the Polycomb group of epigenetic regulators. However, despite a strong correlation between replication-independent histones and active genes, we find that replication-independent H3K27 is not required for gene activation. As mutations in replication-independent H3K27 have recently been identified in human cancers, this work may help to inform the mechanisms by which histone mutations contribute to human disease.
Collapse
Affiliation(s)
- Mary Leatham-Jensen
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Christopher M. Uyehara
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Brian D. Strahl
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - A. Gregory Matera
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Robert J. Duronio
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Daniel J. McKay
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
- * E-mail:
| |
Collapse
|
168
|
Zhang Y, Mi W, Xue Y, Shi X, Kutateladze TG. The ZZ domain as a new epigenetic reader and a degradation signal sensor. Crit Rev Biochem Mol Biol 2019; 54:1-10. [PMID: 30691308 DOI: 10.1080/10409238.2018.1564730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although relatively small in size, the ZZ-type zinc finger (ZZ) domain is a versatile signaling module that is implicated in a diverse set of cell signaling events. Here, we highlight the most recent studies focused on the ZZ domain function as a histone reader and a sensor of protein degradation signals. We review and compare the molecular and structural mechanisms underlying targeting the amino-terminal sequences of histone H3 and arginylated substrates by the ZZ domain. We also discuss the ZZ domain sensitivity to histone PTMs and summarize biological outcomes associated with the recognition of histone and non-histone ligands by the ZZ domain-containing proteins and complexes.
Collapse
Affiliation(s)
- Yi Zhang
- a Department of Pharmacology , University of Colorado School of Medicine , Aurora , CO , USA
| | - Wenyi Mi
- b Center for Epigenetics Van Andel Research Institute , Grand Rapids , MI , USA
| | - Yongming Xue
- c Genetics and Epigenetics Graduate Program , The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences , Houston , TX , USA
| | - Xiaobing Shi
- b Center for Epigenetics Van Andel Research Institute , Grand Rapids , MI , USA
| | - Tatiana G Kutateladze
- a Department of Pharmacology , University of Colorado School of Medicine , Aurora , CO , USA
| |
Collapse
|
169
|
Samee MAH, Bruneau BG, Pollard KS. A De Novo Shape Motif Discovery Algorithm Reveals Preferences of Transcription Factors for DNA Shape Beyond Sequence Motifs. Cell Syst 2019; 8:27-42.e6. [PMID: 30660610 PMCID: PMC6368855 DOI: 10.1016/j.cels.2018.12.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 08/18/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
Abstract
DNA shape adds specificity to sequence motifs but has not been explored systematically outside this context. We hypothesized that DNA-binding proteins (DBPs) preferentially occupy DNA with specific structures ("shape motifs") regardless of whether or not these correspond to high information content sequence motifs. We present ShapeMF, a Gibbs sampling algorithm that identifies de novo shape motifs. Using binding data from hundreds of in vivo and in vitro experiments, we show that most DBPs have shape motifs and can occupy these in the absence of sequence motifs. This "shape-only binding" is common for many DBPs and in regions co-bound by multiple DBPs. When shape and sequence motifs co-occur, they can be overlapping, flanking, or separated by consistent spacing. Finally, DBPs within the same protein family have different shape motifs, explaining their distinct genome-wide occupancy despite having similar sequence motifs. These results suggest that shape motifs not only complement sequence motifs but also facilitate recognition of DNA beyond conventionally defined sequence motifs.
Collapse
Affiliation(s)
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Epidemiology & Biostatistics, Institute for Human Genetics, Quantitative Biology Institute, and Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
170
|
Curtis NL, Bolanos-Garcia VM. The Anaphase Promoting Complex/Cyclosome (APC/C): A Versatile E3 Ubiquitin Ligase. Subcell Biochem 2019; 93:539-623. [PMID: 31939164 DOI: 10.1007/978-3-030-28151-9_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
In the present chapter we discuss the essential roles of the human E3 ubiquitin ligase Anaphase Promoting Complex/Cyclosome (APC/C) in mitosis as well as the emerging evidence of important APC/C roles in cellular processes beyond cell division control such as regulation of genomic integrity and cell differentiation of the nervous system. We consider the potential incipient role of APC/C dysregulation in the pathophysiology of the neurological disorder Alzheimer's disease (AD). We also discuss how certain Deoxyribonucleic Acid (DNA) and Ribonucleic Acid (RNA) viruses take control of the host's cell division regulatory system through harnessing APC/C ubiquitin ligase activity and hypothesise the plausible molecular mechanisms underpinning virus manipulation of the APC/C. We also examine how defects in the function of this multisubunit protein assembly drive abnormal cell proliferation and lastly argue the potential of APC/C as a promising therapeutic target for the development of innovative therapies for the treatment of chronic malignancies such as cancer.
Collapse
Affiliation(s)
- Natalie L Curtis
- Faculty of Health and Life Sciences, Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, England, UK
| | - Victor M Bolanos-Garcia
- Faculty of Health and Life Sciences, Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, England, UK.
| |
Collapse
|
171
|
Biswas S, Chen S, Liang G, Feng B, Cai L, Khan ZA, Chakrabarti S. Curcumin Analogs Reduce Stress and Inflammation Indices in Experimental Models of Diabetes. Front Endocrinol (Lausanne) 2019; 10:887. [PMID: 31920992 PMCID: PMC6930691 DOI: 10.3389/fendo.2019.00887] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation and oxidative stress lead to a multitude of adverse cellular responses in target organs of chronic diabetic complications. Curcumin, a highly investigated phytochemical, has been shown to exhibit both anti-inflammatory and antioxidant activities. However, the clinical application of curcumin has been greatly limited due to a poor pharmacokinetic profile. To overcome these limitations, we have generated analogs of curcumin to enhance bioavailability and offer a preferable pharmacokinetic profile. Here, we explored the effects of two mono-carbonyl curcumin analogs, L2H21 and L50H46, in alleviating indices of inflammation and oxidative stress in cell culture and mouse model of diabetic complications. Our results show that L2H21 and L50H46 normalize inflammatory mediators (IL-6 and TNF-α), extracellular matrix proteins (FN and COL4α1), vasoactive factors (VEGF and ET-1) and a key transcriptional coactivator (p300) in cultured human retinal microvascular endothelial cells (HRECs) and dermal-derived microvascular endothelial cells (HMVECs) challenged with high levels of glucose. These curcumin analogs also reduced glucose-induced oxidative DNA damage as evidenced by 8-OHdG labeling. We further show that treatment of streptozotocin-induced diabetic mice with curcumin analogs prevents cardiac and renal dysfunction. The preservation of target tissue function was associated with normalization of pro-inflammatory cytokines and matrix proteins. Collectively, our results show that L2H21 and L50H46 offer the anti-inflammatory and antioxidant activities as has been reported for curcumin, and may provide a clinically applicable therapeutic option for the treatment of diabetic complications.
Collapse
Affiliation(s)
- Saumik Biswas
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Shali Chen
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Guang Liang
- Chemical Biology Research Centre, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Biao Feng
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, United States
| | - Zia A. Khan
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- *Correspondence: Zia A. Khan
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- Subrata Chakrabarti
| |
Collapse
|
172
|
Abstract
Posttranslational acetylation modifications of proteins have important consequences for cell biology, including effects on protein trafficking and cellular localization as well as on the interactions of acetylated proteins with other proteins and macromolecules such as DNA. Experiments to uncover and characterize protein acetylation events have historically been more challenging than investigating another common posttranslational modification, protein phosphorylation. More recently, high-quality antibodies that recognize acetylated lysine residues present in acetylated proteins and improved proteomic methodologies have facilitated the discovery that acetylation occurs on numerous cellular proteins and allowed characterization of the dynamics and functional effects of many acetylation events. This article summarizes some established biochemical information about how protein acetylation takes place and is regulated, in order to lay the foundation for subsequent descriptions of strategies used by our lab and others either to directly study acetylation of an individual factor or to identify groups of proteins targeted for acetylation that can then be examined in isolation.
Collapse
Affiliation(s)
- David K Orren
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA.
| | - Amrita Machwe
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
173
|
Nassar AH, Umeton R, Kim J, Lundgren K, Harshman L, Van Allen EM, Preston M, Dong F, Bellmunt J, Mouw KW, Choueiri TK, Sonpavde G, Kwiatkowski DJ. Mutational Analysis of 472 Urothelial Carcinoma Across Grades and Anatomic Sites. Clin Cancer Res 2018; 25:2458-2470. [PMID: 30593515 DOI: 10.1158/1078-0432.ccr-18-3147] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/17/2018] [Accepted: 12/21/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of this study is to characterize the mutational landscape across the spectrum of urothelial carcinoma (UC) to identify mutational features and potential therapeutic targets. EXPERIMENTAL DESIGN Using targeted exome sequencing (n = 237 genes), we analyzed the mutation spectra of 82 low-grade nonmuscle-invasive bladder cancers (LG-NMIBC), 126 high-grade (HG) NMIBC, 199 muscle-invasive bladder cancers (MIBC), 10 LG-upper tract urothelial cancers (LG-UTUC), and 55 HG-UTUC. RESULTS FGFR3 and KDM6A mutations were significantly more common in LG-NMIBC (72% and 44%, respectively) versus other bladder subtypes. FGFR3 alterations were also enriched in LG-UTUC versus HG-UTUC tumors (80% vs. 16%). In contrast, TP53 and RB1 mutations were significantly more frequent in all 3 HG urothelial carcinoma subtypes than in LG-NIMBC (45%-58% vs. 4%; 9%-22% vs. 0; respectively). Among LG-NMIBC tumors, KDM6A mutations were more common in women than in men (71% vs. 38%). HG-NMIBC and MIBC had higher tumor mutational burden (TMB) than LG-NMIBC (P = 0.001 and P < 0.01, respectively). DNA-damage repair (DDR) alterations were associated with a higher TMB in HG-NMIBC and MIBC tumors, and these two tumor types were also enriched for an APOBEC mutational signature compared with LG-NMIBC and HG-UTUC. Alterations in FGFR3, PIK3CA, and EP300 correlated with worse overall survival in HG-UTUC and occurred concurrently. CONCLUSIONS Our analysis suggests that a fraction of MIBCs likely arise from precursor lesions other than LG-NMIBC. KDM6A mutations are twice as common in women with LG-NIMBC than those in men. DDR gene mutations and APOBEC mutagenesis drive mutations in HG-NMIBC and MIBC. UTUC has a distinct mutation profile from bladder cancer.
Collapse
Affiliation(s)
- Amin H Nassar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Renato Umeton
- Department of Informatics, Dana-Farber Cancer Institute, Boston, Massachusetts.,Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jaegil Kim
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Kevin Lundgren
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lauren Harshman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Mark Preston
- Department of Urology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joaquim Bellmunt
- Department of Medical Oncology, IMIM-Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Guru Sonpavde
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David J Kwiatkowski
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
174
|
Roy A, Palli SR. Epigenetic modifications acetylation and deacetylation play important roles in juvenile hormone action. BMC Genomics 2018; 19:934. [PMID: 30547764 PMCID: PMC6295036 DOI: 10.1186/s12864-018-5323-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Epigenetic modifications including DNA methylation and post-translational modifications of histones are known to regulate gene expression. Antagonistic activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs) mediate transcriptional reprogramming during insect development as shown in Drosophila melanogaster and other insects. Juvenile hormones (JH) play vital roles in the regulation of growth, development, metamorphosis, reproduction and other physiological processes. However, our current understanding of epigenetic regulation of JH action is still limited. Hence, we studied the role of CREB binding protein (CBP, contains HAT domain) and Trichostatin A (TSA, HDAC inhibitor) on JH action. RESULTS Exposure of Tribolium castaneum cells (TcA cells) to JH or TSA caused an increase in expression of Kr-h1 (a known JH-response gene) and 31 or 698 other genes respectively. Knockdown of the gene coding for CBP caused a decrease in the expression of 456 genes including Kr-h1. Interestingly, the expression of several genes coding for transcription factors, nuclear receptors, P450 and fatty acid synthase family members that are known to mediate JH action were affected by CBP knockdown or TSA treatment. CONCLUSIONS These data suggest that acetylation and deacetylation mediated by HATs and HDACs play an important role in JH action.
Collapse
Affiliation(s)
- Amit Roy
- Department of Entomology, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546 USA
- Faculty of Forestry and Wood Sciences, EXTEMIT-K, Czech University of Life Sciences, Kamýcká 1176, Prague 6, 165 21 Suchdol, Czech Republic
| | - Subba Reddy Palli
- Department of Entomology, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546 USA
| |
Collapse
|
175
|
Friedrich J, Heim L, Trufa DI, Sirbu H, Rieker RJ, Chiriac MT, Finotto S. STAT1 deficiency supports PD-1/PD-L1 signaling resulting in dysfunctional TNFα mediated immune responses in a model of NSCLC. Oncotarget 2018; 9:37157-37172. [PMID: 30647851 PMCID: PMC6324686 DOI: 10.18632/oncotarget.26441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/26/2018] [Indexed: 12/27/2022] Open
Abstract
In this study we described that Signal Transducer and Activator of Transcription 1 (STAT1) is a key point regulator of PD-1 in tumour infiltrating lymphocytes and PD-L1 in Tumour associated macrophages (TAM) in NSCLC. In our murine model of adenocarcinoma targeted deletion of Stat1 was found associated with enhanced tumour growth, impaired differentiation into M1-like macrophages from the bone marrow, the accumulation of tumor associated macrophages overexpressing PD-L1 and impaired T cell responses in the tumor microenvironment by affecting TNFα responses. In our human NSCLC patient cohort we found that loss of isoforms STAT1 α and STAT1β mRNA in the tumoural region of the lung correlates with increased tumor size in NSCLC patients. Therefore, STAT1 isoform regulation could be considered for future therapeutical strategies associated to current immune-checkpoint blockade therapy in NSCLC.
Collapse
Affiliation(s)
- Juliane Friedrich
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lisanne Heim
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Mircea T Chiriac
- Department of Medicine 1-Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
176
|
Chen W, Nyuydzefe MS, Weiss JM, Zhang J, Waksal SD, Zanin-Zhorov A. ROCK2, but not ROCK1 interacts with phosphorylated STAT3 and co-occupies TH17/TFH gene promoters in TH17-activated human T cells. Sci Rep 2018; 8:16636. [PMID: 30413785 PMCID: PMC6226480 DOI: 10.1038/s41598-018-35109-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/30/2018] [Indexed: 02/04/2023] Open
Abstract
Rho-associated coiled-coil kinase (ROCK)2 targeting down-regulates autoimmune responses in animal models and patients, however the underlying molecular mechanism is still an enigma. We report that ROCK2 binds phosphorylated-STAT3 and its kinase activity controls the formation of ROCK2/STAT3/JAK2 complex and optimal STAT3 phosphorylation in human CD4+ T cells during T helper 17 (TH17)-skewing. Moreover, chromatin-immunoprecipitation sequencing (ChIP-seq) analysis revealed that, genome-wide, about 70% of ROCK2 and STAT3 peaks overlapped and co-localized to several key genes controlling TH17 and T follicular helper (TFH) cell functions. Specifically, the co-occupancy of ROCK2 and STAT3 on the Irf4 and Bcl6 genes was validated by ChIP-qPCR analysis. Furthermore, the binding of ROCK2 to both the Irf4 and Bcl6 promoters was attenuated by STAT3 silencing as well as by selective ROCK2 inhibitor. Thus, the present study demonstrated previously unidentified evidence that ROCK2-mediated signaling in the cytosol provides a positive feed-forward signal for nuclear ROCK2 to be recruited to the chromatin by STAT3 and potentially regulates TH17/TFH gene transcription.
Collapse
Affiliation(s)
- Wei Chen
- Kadmon Corporation, LLC, New York, NY, 10016, USA
| | | | | | - Jingya Zhang
- Kadmon Corporation, LLC, New York, NY, 10016, USA
| | - Samuel D Waksal
- Kadmon Corporation, LLC, New York, NY, 10016, USA.,Current Weill Cornell Medicine, New York, NY, 10021, USA
| | | |
Collapse
|
177
|
Wnt/β-Catenin Signaling as a Potential Target for the Treatment of Liver Cirrhosis Using Antifibrotic Drugs. Int J Mol Sci 2018; 19:ijms19103103. [PMID: 30308992 PMCID: PMC6213128 DOI: 10.3390/ijms19103103] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
Abstract
Cirrhosis is a form of liver fibrosis resulting from chronic hepatitis and caused by various liver diseases, including viral hepatitis, alcoholic liver damage, nonalcoholic steatohepatitis, and autoimmune liver disease. Cirrhosis leads to various complications, resulting in poor prognoses; therefore, it is important to develop novel antifibrotic therapies to counter liver cirrhosis. Wnt/β-catenin signaling is associated with the development of tissue fibrosis, making it a major therapeutic target for treating liver fibrosis. In this review, we present recent insights into the correlation between Wnt/β-catenin signaling and liver fibrosis and discuss the antifibrotic effects of the cAMP-response element binding protein/β-catenin inhibitor PRI-724.
Collapse
|
178
|
Zhang Y, Zheng D, Zhou T, Song H, Hulsurkar M, Su N, Liu Y, Wang Z, Shao L, Ittmann M, Gleave M, Han H, Xu F, Liao W, Wang H, Li W. Androgen deprivation promotes neuroendocrine differentiation and angiogenesis through CREB-EZH2-TSP1 pathway in prostate cancers. Nat Commun 2018; 9:4080. [PMID: 30287808 PMCID: PMC6172226 DOI: 10.1038/s41467-018-06177-2] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/20/2018] [Indexed: 01/19/2023] Open
Abstract
The incidence of aggressive neuroendocrine prostate cancers (NEPC) related to androgen-deprivation therapy (ADT) is rising. NEPC is still poorly understood, such as its neuroendocrine differentiation (NED) and angiogenic phenotypes. Here we reveal that NED and angiogenesis are molecularly connected through EZH2 (enhancer of zeste homolog 2). NED and angiogenesis are both regulated by ADT-activated CREB (cAMP response element-binding protein) that in turn enhances EZH2 activity. We also uncover anti-angiogenic factor TSP1 (thrombospondin-1, THBS1) as a direct target of EZH2 epigenetic repression. TSP1 is downregulated in advanced prostate cancer patient samples and negatively correlates with NE markers and EZH2. Furthermore, castration activates the CREB/EZH2 axis, concordantly affecting TSP1, angiogenesis and NE phenotypes in tumor xenografts. Notably, repressing CREB inhibits the CREB/EZH2 axis, tumor growth, NED, and angiogenesis in vivo. Taken together, we elucidate a new critical pathway, consisting of CREB/EZH2/TSP1, underlying ADT-enhanced NED and angiogenesis during prostate cancer progression.
Collapse
Affiliation(s)
- Yan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Anesthesiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dayong Zheng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510513, China
| | - Ting Zhou
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, 201400, China
| | - Haiping Song
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Breast and Thyroid Surgery Center, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mohit Hulsurkar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Ning Su
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Oncology, Guangzhou Chest Hospital, Guangzhou, 510095, China
| | - Ying Liu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Pathology, Xiangya Hospital and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
| | - Zheng Wang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Long Shao
- Department of Pathology and Immunology, Baylor College of Medicine, and Michael E. DeBakey VAMC, Houston, TX 77030, USA
| | - Michael Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, and Michael E. DeBakey VAMC, Houston, TX 77030, USA
| | - Martin Gleave
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Huanxing Han
- Department of Pharmacy, Changzheng Hospital, Shanghai, 200003, China
| | - Feng Xu
- Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, 201400, China
| | - Wangjun Liao
- Department of Medical Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongbo Wang
- Department of Gynaecology and Obstetrics, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenliang Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
- Division of Oncology, Department of Internal Medicine, and Memorial Herman Cancer Center, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
179
|
Haldar R, Shaashua L, Lavon H, Lyons YA, Zmora O, Sharon E, Birnbaum Y, Allweis T, Sood AK, Barshack I, Cole S, Ben-Eliyahu S. Perioperative inhibition of β-adrenergic and COX2 signaling in a clinical trial in breast cancer patients improves tumor Ki-67 expression, serum cytokine levels, and PBMCs transcriptome. Brain Behav Immun 2018; 73:294-309. [PMID: 29800703 DOI: 10.1016/j.bbi.2018.05.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Catecholamines and prostaglandins are secreted abundantly during the perioperative period in response to stress and surgery, and were shown by translational studies to promote tumor metastasis. Here, in a phase-II biomarker clinical trial in breast cancer patients (n = 38), we tested the combined perioperative use of the β-blocker, propranolol, and the COX2-inhibitor, etodolac, scheduled for 11 consecutive perioperative days, starting 5 days before surgery. Blood samples were taken before treatment (T1), on the mornings before and after surgery (T2&T3), and after treatment cessation (T4). Drugs were well tolerated. Results based on a-priori hypotheses indicated that already before surgery (T2), serum levels of pro-inflammatory IL-6, CRP, and IFNγ, and anti-inflammatory, cortisol and IL-10, increased. At T2 and/or T3, drug treatment reduced serum levels of the above pro-inflammatory cytokines and of TRAIL, as well as activity of multiple inflammation-related transcription factors (including NFκB, STAT3, ISRE), but not serum levels of cortisol, IL-10, IL-18, IL-8, VEGF and TNFα. In the excised tumor, treatment reduced the expression of the proliferation marker Ki-67, and positively affected its transcription factors SP1 and AhR. Exploratory analyses of transcriptome modulation in PBMCs revealed treatment-induced improvement at T2/T3 in several transcription factors that in primary tumors indicate poor prognosis (CUX1, THRa, EVI1, RORa, PBX1, and T3R), angiogenesis (YY1), EMT (GATA1 and deltaEF1/ZEB1), proliferation (GATA2), and glucocorticoids response (GRE), while increasing the activity of the oncogenes c-MYB and N-MYC. Overall, the drug treatment may benefit breast cancer patients through reducing systemic inflammation and pro-metastatic/pro-growth biomarkers in the excised tumor and PBMCs.
Collapse
Affiliation(s)
- Rita Haldar
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Lee Shaashua
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Hagar Lavon
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Yasmin A Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Oded Zmora
- Department of Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Yehudit Birnbaum
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Tanir Allweis
- Department of Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Steve Cole
- Department of Medicine, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel.
| |
Collapse
|
180
|
Wang Q, An X, Xu J, Wang Y, Liu L, Leung ELH, Yao X. Classical molecular dynamics and metadynamics simulations decipher the mechanism of CBP30 selectively inhibiting CBP/p300 bromodomains. Org Biomol Chem 2018; 16:6521-6530. [PMID: 30160288 DOI: 10.1039/c8ob01526k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The selective modulation of individual bromodomains (BDs) by small molecules represents an important strategy for the treatment of various cancers, considering that the BD-containing proteins share common BD structures and distinct pharmacological functions. Small molecule inhibitors targeting BDs outside of the bromodomain and extraterminal domain (BET, including BRD2-4 and BRDT) family are particularly lacking. CBP30 exhibited excellent selectivity for the transcriptional coactivators CBP (CREB binding protein) and p300 bromodomains, providing a new opportunity for designing selective non-BET inhibitors. Here, we performed classical molecular dynamics (cMD) and metadynamics simulations to reveal the selective mechanism of CBP30 binding with CBP/p300 and BRD4-BD1/BD2 bromodomains. The cMD simulations combined with binding free energy calculations were performed to compare the overall features of CBP30 binding with CBP/p300 and BRD4-BD1/BD2 bromodomains. Arg1173/1137, as the unique residue for CBP/p300, was responsible for the selective binding to CBP30 via cation-π and hydrogen bond interactions. Metadynamics simulation, together with unbinding free energy profiles, suggested that the dissociation pathways of CBP30 from CBP/p300 and BRD4-BD1/BD2 bromodomains were different, with the unbinding of the former being more difficult. These findings will be helpful for novel CBP/p300-inhibitor design and rational structural modification of existing inhibitors to increase their selectivity.
Collapse
Affiliation(s)
- Qianqian Wang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China.
| | | | | | | | | | | | | |
Collapse
|
181
|
Zhang Y, Xue Y, Shi J, Ahn J, Mi W, Ali M, Wang X, Klein BJ, Wen H, Li W, Shi X, Kutateladze TG. The ZZ domain of p300 mediates specificity of the adjacent HAT domain for histone H3. Nat Struct Mol Biol 2018; 25:841-849. [PMID: 30150647 PMCID: PMC6482957 DOI: 10.1038/s41594-018-0114-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/18/2018] [Indexed: 12/24/2022]
Abstract
Human p300 is a transcriptional co-activator and a major acetyltransferase that acetylates histones and other proteins facilitating gene transcription. The activity of p300 relies on the fine-tuned interactome that involves a dozen p300 domains and hundreds of binding partners and links p300 to a wide range of vital signaling events. Here, we report on a novel function of the ZZ-type zinc finger (ZZ) of p300 as a reader of histone H3. We show that the ZZ domain and acetyllysine recognizing bromodomain (BD) of p300 play critical roles in modulating p300 enzymatic activity and its association with chromatin. Acetyllysine binding of BD is essential for acetylation of histones H3 and H4, whereas interaction of the ZZ domain with H3 promotes selective acetylation of histone H3K27 and H3K18.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yongming Xue
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jiejun Shi
- Dan L. Duncan Cancer Center, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - JaeWoo Ahn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wenyi Mi
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Muzaffar Ali
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xiaolu Wang
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Brianna J Klein
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hong Wen
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Wei Li
- Dan L. Duncan Cancer Center, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaobing Shi
- Department of Epigenetics and Molecular Carcinogenesis, Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Genetics and Epigenetics Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA. .,Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA.
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
182
|
Wang J, Qiu Z, Wu Y. Ubiquitin Regulation: The Histone Modifying Enzyme's Story. Cells 2018; 7:cells7090118. [PMID: 30150556 PMCID: PMC6162602 DOI: 10.3390/cells7090118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
Histone post-translational modifications influence many fundamental cellular events by regulating chromatin structure and gene transcriptional activity. These modifications are highly dynamic and tightly controlled, with many enzymes devoted to the addition and removal of these modifications. Interestingly, these modifying enzymes are themselves fine-tuned and precisely regulated at the level of protein turnover by ubiquitin-proteasomal processing. Here, we focus on recent progress centered on the mechanisms regulating ubiquitination of histone modifying enzymes, including ubiquitin proteasomal degradation and the reverse process of deubiquitination. We will also discuss the potential pathophysiological significance of these processes.
Collapse
Affiliation(s)
- Jianlin Wang
- Department of Pharmacology & Nutritional Sciences, University of Kentucky School of Medicine, KY 40506, USA.
- Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA.
| | - Zhaoping Qiu
- Department of Pharmacology & Nutritional Sciences, University of Kentucky School of Medicine, KY 40506, USA.
- Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA.
| | - Yadi Wu
- Department of Pharmacology & Nutritional Sciences, University of Kentucky School of Medicine, KY 40506, USA.
- Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA.
| |
Collapse
|
183
|
Bemanian V, Noone JC, Sauer T, Touma J, Vetvik K, Søderberg-Naucler C, Lindstrøm JC, Bukholm IR, Kristensen VN, Geisler J. Somatic EP300-G211S mutations are associated with overall somatic mutational patterns and breast cancer specific survival in triple-negative breast cancer. Breast Cancer Res Treat 2018; 172:339-351. [PMID: 30132219 DOI: 10.1007/s10549-018-4927-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE We have compared the mutational profiles of human breast cancer tumor samples belonging to all major subgroups with special emphasis on triple-negative breast cancer (TNBC). Our major goal was to identify specific mutations that could be potentially used for clinical decision making in TNBC patients. PATIENTS AND METHODS Primary tumor specimens from 149 Norwegian breast cancer patients were available. We analyzed the tissue samples for somatic mutations in 44 relevant breast cancer genes by targeted next-generation sequencing. As a second confirmatory technique, we performed pyrosequencing on selected samples. RESULTS We observed a distinct subgroup of TNBC patients, characterized by an almost completely lack of pathogenic somatic mutations. A point mutation in the adenoviral E1A binding protein p300 (EP300-G211S) was significantly correlated to this TNBC subgroup. The EP300-G211S mutation was exclusively found in the TNBC patients and its presence reduced the chance for other pathological somatic mutations in typical breast cancer genes investigated in our gene panel by 94.9% (P < 0.005). Interestingly, the EP300-G211S mutation also predicted a lower risk for relapses and decreased breast cancer-specific mortality during long-term follow-up of the patients. CONCLUSION Next-generation sequencing revealed specific mutations in EP300 to be associated with the mutational patterns in typical breast cancer genes and long-term outcome of triple-negative breast cancer patients.
Collapse
Affiliation(s)
- Vahid Bemanian
- Section of Gene Technology, Akershus University Hospital, 1478, Lørenskog, Norway
| | | | - Torill Sauer
- Department of Pathology, Akershus University Hospital, 1478, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway
| | - Joel Touma
- Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway.,Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Katja Vetvik
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Cecilia Søderberg-Naucler
- Department of Medicine at Solna, Experimental Cardiovascular Research Unit and Departments of Medicine and Neurology, Center for Molecular Medicine, Karolinska Institute, 17176, Stockholm, Sweden
| | - Jonas Christoffer Lindstrøm
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Health Services Research Unit, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Ida Rashida Bukholm
- Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway.,Norwegian System of Compensation to Patients, Oslo, Norway.,The Norwegian University of Life Sciences, Ås, Norway
| | - Vessela N Kristensen
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Clinical Molecular Biology (EPIGEN), Akershus University Hospital, 1478, Lørenskog, Norway
| | - Jürgen Geisler
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway. .,Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway.
| |
Collapse
|
184
|
Kaypee S, Sahadevan SA, Sudarshan D, Halder Sinha S, Patil S, Senapati P, Kodaganur GS, Mohiyuddin A, Dasgupta D, Kundu TK. Oligomers of human histone chaperone NPM1 alter p300/KAT3B folding to induce autoacetylation. Biochim Biophys Acta Gen Subj 2018; 1862:1729-1741. [DOI: 10.1016/j.bbagen.2018.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 11/27/2022]
|
185
|
Khilji S, Hamed M, Chen J, Li Q. Loci-specific histone acetylation profiles associated with transcriptional coactivator p300 during early myoblast differentiation. Epigenetics 2018; 13:642-654. [PMID: 29927685 PMCID: PMC6140897 DOI: 10.1080/15592294.2018.1489659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Molecular regulation of stem cell differentiation is exerted through both genetic and epigenetic determinants over distal regulatory or enhancer regions. Understanding the mechanistic action of active or poised enhancers is therefore imperative for control of stem cell differentiation. Based on the genome-wide co-occurrence of different epigenetic marks in committed proliferating myoblasts, we have previously generated a 14-state chromatin state model to profile rexinoid-responsive histone acetylation in early myoblast differentiation. Here, we delineate the functional mode of transcription regulators during early myogenic differentiation using genome-wide chromatin state association. We define a role of transcriptional coactivator p300, when recruited by muscle master regulator MyoD, in the establishment and regulation of myogenic loci at the onset of myoblast differentiation. In addition, we reveal an enrichment of loci-specific histone acetylation at p300 associated active or poised enhancers, particularly when enlisted by MyoD. We provide novel molecular insights into the regulation of myogenic enhancers by p300 in concert with MyoD. Our studies present a valuable aptitude for driving condition-specific chromatin state or enhancers pharmacologically to treat muscle-related diseases and for the identification of additional myogenic targets and molecular interactions for therapeutic development. Abbreviations: MRF: Muscle regulatory factor; HAT: Histone acetyltransferase; CBP: CREB-binding protein; ES: Embryonic stem; ATCC: American type culture collection; DM: Differentiation medium; DMEM: Dulbecco’s Modified Eagle Medium; GM: Growth medium; GO: Gene ontology; GREAT: Genomic regions enrichment of annotations tool; FPKM: Fragments per kilobase of transcript per million; GEO: Gene expression omnibus; MACS: Model-based analysis for ChIP-seq
Collapse
Affiliation(s)
- Saadia Khilji
- a Department of Cellular and Molecular Medicine, Faculty of Medicine , University of Ottawa , Ottawa , Ontario , Canada
| | - Munerah Hamed
- a Department of Cellular and Molecular Medicine, Faculty of Medicine , University of Ottawa , Ottawa , Ontario , Canada
| | - Jihong Chen
- b Department of Pathology and Laboratory Medicine, Faculty of Medicine , University of Ottawa , Ottawa , Ontario , Canada
| | - Qiao Li
- a Department of Cellular and Molecular Medicine, Faculty of Medicine , University of Ottawa , Ottawa , Ontario , Canada.,b Department of Pathology and Laboratory Medicine, Faculty of Medicine , University of Ottawa , Ottawa , Ontario , Canada
| |
Collapse
|
186
|
Li J, Tang H, Kurtán T, Mándi A, Zhuang CL, Su L, Zheng GL, Zhang W. Swinhoeisterols from the South China Sea Sponge Theonella swinhoei. JOURNAL OF NATURAL PRODUCTS 2018; 81:1645-1650. [PMID: 29989811 DOI: 10.1021/acs.jnatprod.8b00281] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Swinhoeisterols C-F (1-4), four new steroids having a rearranged 6/6/5/7 ring system, were isolated from the Xisha sponge Theonella swinhoei, together with the known analogue swinhoeisterol A (5). Their structures were determined based on spectroscopic analysis, TDDFT-ECD and optical rotation calculations, and biogenetic correlations. In an in vitro assay, compound 1 showed an inhibitory effect on (h)p300 with an IC50 value of 8.8 μM, whereas compounds 2-4 were not active.
Collapse
Affiliation(s)
- Jiao Li
- School of Pharmacy , Second Military Medical University , 325 Guo-He Road , Shanghai 200433 , People's Republic of China
| | - Hua Tang
- School of Pharmacy , Second Military Medical University , 325 Guo-He Road , Shanghai 200433 , People's Republic of China
| | - Tibor Kurtán
- Department of Organic Chemistry , University of Debrecen , POB 400, H-4002 Debrecen , Hungary
| | - Attila Mándi
- Department of Organic Chemistry , University of Debrecen , POB 400, H-4002 Debrecen , Hungary
| | - Chun-Lin Zhuang
- School of Pharmacy , Second Military Medical University , 325 Guo-He Road , Shanghai 200433 , People's Republic of China
| | - Li Su
- School of Pharmacy , Second Military Medical University , 325 Guo-He Road , Shanghai 200433 , People's Republic of China
| | - Gui-Liang Zheng
- Department of Otorhinolaryngology, Head and Neck Surgery , Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , 1665 Kong-Jiang Road , Shanghai 200092 , People's Republic of China
| | - Wen Zhang
- School of Pharmacy , Second Military Medical University , 325 Guo-He Road , Shanghai 200433 , People's Republic of China
| |
Collapse
|
187
|
Tamošaitis L, Smales CM. Meta-Analysis of Publicly Available Chinese Hamster Ovary (CHO) Cell Transcriptomic Datasets for Identifying Engineering Targets to Enhance Recombinant Protein Yields. Biotechnol J 2018; 13:e1800066. [DOI: 10.1002/biot.201800066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/23/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Linas Tamošaitis
- Industrial Biotechnology Centre and School of Biosciences; University of Kent; Canterbury Kent CT2 7NJ UK
| | - Christopher Mark Smales
- Industrial Biotechnology Centre and School of Biosciences; University of Kent; Canterbury Kent CT2 7NJ UK
| |
Collapse
|
188
|
|
189
|
Dou C, Liu Z, Tu K, Zhang H, Chen C, Yaqoob U, Wang Y, Wen J, van Deursen J, Sicard D, Tschumperlin D, Zou H, Huang WC, Urrutia R, Shah VH, Kang N. P300 Acetyltransferase Mediates Stiffness-Induced Activation of Hepatic Stellate Cells Into Tumor-Promoting Myofibroblasts. Gastroenterology 2018; 154:2209-2221.e14. [PMID: 29454793 PMCID: PMC6039101 DOI: 10.1053/j.gastro.2018.02.015] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) contribute to desmoplasia and stiffness of liver metastases by differentiating into matrix-producing myofibroblasts. We investigated whether stiffness due to the presence of tumors increases activation of HSCs into myofibroblasts and their tumor-promoting effects, as well as the role of E1A binding protein p300, a histone acetyltransferase that regulates transcription, in these processes. METHODS HSCs were isolated from liver tissues of patients, mice in which the p300 gene was flanked by 2 loxP sites (p300F/F mice), and p300+/+ mice (controls). The HSCs were placed on polyacrylamide gels with precisely defined stiffness, and their activation (differentiation into myofibroblasts) was assessed by immunofluorescence and immunoblot analyses for alpha-smooth muscle actin. In HSCs from mice, the p300 gene was disrupted by cre recombinase. In human HSCs, levels of p300 were knocked down with small hairpin RNAs or a mutant form of p300 that is not phosphorylated by AKT (p300S1834A) was overexpressed. Human HSCs were also cultured with inhibitors of p300 (C646), PI3K signaling to AKT (LY294002), or RHOA (C3 transferase) and effects on stiffness-induced activation were measured. RNA sequencing and chromatin immunoprecipitation-quantitative polymerase chain reaction were used to identify HSC genes that changed expression levels in response to stiffness. We measured effects of HSC-conditioned media on proliferation of HT29 colon cancer cells and growth of tumors following subcutaneous injection of these cells into mice. MC38 colon cancer cells were injected into portal veins of p300F/Fcre and control mice, and liver metastases were measured. p300F/Fcre and control mice were given intraperitoneal injections of CCl4 to induce liver fibrosis. Liver tissues were collected and analyzed by immunofluorescence, immunoblot, and histology. RESULTS Substrate stiffness was sufficient to activate HSCs, leading to nuclear accumulation of p300. Disrupting p300 level or activity blocked stiffness-induced activation of HSCs. In HSCs, substrate stiffness activated AKT signaling via RHOA to induce phosphorylation of p300 at serine 1834; this caused p300 to translocate to the nucleus, where it up-regulated transcription of genes that increase activation of HSCs and metastasis, including CXCL12. MC38 cells, injected into portal veins, formed fewer metastases in livers of p300F/Fcre mice than control mice. Expression of p300 was increased in livers of mice following injection of CCl4; HSC activation and collagen deposition were reduced in livers of p300F/Fcre mice compared with control mice. CONCLUSIONS In studies of mice, we found liver stiffness to activate HSC differentiation into myofibroblasts, which required nuclear accumulation of p300. p300 increases HSC expression of genes that promote metastasis.
Collapse
Affiliation(s)
- Changwei Dou
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,Department of Hepatobiliary Surgery, Zhejiang provincial People's Hospital, Hangzhou, China
| | - Zhikui Liu
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, 1st Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China,Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Hongbin Zhang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Chen Chen
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Usman Yaqoob
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN
| | - Yuanguo Wang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Jialing Wen
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota
| | - Jan van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Daniel Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hongzhi Zou
- Guangdong Institute of Gastroenterology, 6th Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Science, China Medical University, Taiwan, R.O.C
| | - Raul Urrutia
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN
| | - Vijay H. Shah
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN,To whom correspondence should be addressed: Ningling Kang, Ph.D., Hormel Institute, 801 16th Ave NE Austin MN 55912. Fax: (507) 437-9606. Phone: (507) 437-9680. . Vijay Shah, M.D., Mayo Clinic, 200 1st ST SW Rochester MN 55915. Fax: (507) 255-6318. Phone: (507) 255-6028.
| | - Ningling Kang
- Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
190
|
Pulecio J, Verma N, Mejía-Ramírez E, Huangfu D, Raya A. CRISPR/Cas9-Based Engineering of the Epigenome. Cell Stem Cell 2018; 21:431-447. [PMID: 28985525 DOI: 10.1016/j.stem.2017.09.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Determining causal relationships between distinct chromatin features and gene expression, and ultimately cell behavior, remains a major challenge. Recent developments in targetable epigenome-editing tools enable us to assign direct transcriptional and functional consequences to locus-specific chromatin modifications. This Protocol Review discusses the unprecedented opportunity that CRISPR/Cas9 technology offers for investigating and manipulating the epigenome to facilitate further understanding of stem cell biology and engineering of stem cells for therapeutic applications. We also provide technical considerations for standardization and further improvement of the CRISPR/Cas9-based tools to engineer the epigenome.
Collapse
Affiliation(s)
- Julian Pulecio
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, 3rd floor, Avenue Gran Via 199-203, Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Nipun Verma
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA; Weill Graduate School of Medical Sciences at Cornell University/The Rockefeller University/Sloan Kettering Institute Tri-Institutional M.D.-Ph.D. Program, 1300 York Avenue, New York, NY 10065, USA
| | - Eva Mejía-Ramírez
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, 3rd floor, Avenue Gran Via 199-203, Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA.
| | - Angel Raya
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, 3rd floor, Avenue Gran Via 199-203, Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
191
|
Pasqualucci L, Dalla-Favera R. Genetics of diffuse large B-cell lymphoma. Blood 2018; 131:2307-2319. [PMID: 29666115 PMCID: PMC5969374 DOI: 10.1182/blood-2017-11-764332] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/15/2018] [Indexed: 02/07/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), the most frequent subtype of lymphoid malignancy, remains a significant clinical challenge, as ∼30% of patients are not cured. Over the past decade, remarkable progress has been made in the understanding of the pathogenesis of this disease, spurred by the implementation of powerful genomic technologies that enabled the definition of its genetic and epigenetic landscape. These studies have uncovered a multitude of genomic alterations that contribute to the initiation and maintenance of the tumor clone by disrupting biological functions known to be critical for the normal biology of its cells of origin, germinal center B cells. The identified alterations involve epigenetic remodeling, block of differentiation, escape from immune surveillance, and the constitutive activation of several signal transduction pathways. This wealth of new information offers unique opportunities for the development of improved diagnostic and prognostic tools that could help guide the clinical management of DLBCL patients. Furthermore, a number of the mutated genes identified are potentially actionable targets that are currently being explored for the development of novel therapeutic strategies. This review summarizes current knowledge of the most common genetic alterations associated with DLBCL in relation to their functional impact on the malignant transformation process, and discusses their clinical implications for mechanism-based therapeutics.
Collapse
Affiliation(s)
- Laura Pasqualucci
- Institute for Cancer Genetics
- Department of Pathology and Cell Biology
| | - Riccardo Dalla-Favera
- Institute for Cancer Genetics
- Department of Pathology and Cell Biology
- Department of Genetics, and
- Department of Microbiology and Immunology, Columbia University, New York, NY
| |
Collapse
|
192
|
Gee CT, Arntson KE, Koleski EJ, Staebell RL, Pomerantz WCK. Dual Labeling of the CBP/p300 KIX Domain for 19 F NMR Leads to Identification of a New Small-Molecule Binding Site. Chembiochem 2018; 19:963-969. [PMID: 29430847 PMCID: PMC6251716 DOI: 10.1002/cbic.201700686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Indexed: 12/15/2022]
Abstract
Protein-Observed Fluorine NMR (PrOF NMR) spectroscopy is an emerging technique for screening and characterizing small-molecule-protein interactions. The choice of which amino acid to label for PrOF NMR can be critical for analysis. Here we report the first use of a protein containing two different fluoroaromatic amino acids for NMR studies. Using the KIX domain of the CBP/p300 as a model system, we examine ligand binding of several small-molecule compounds elaborated from our previous fragment screen and identify a new ligand binding site distinct from those used by native transcription factors. This site was further supported by computational modeling (FTMap and Schrödinger) and 1 H,15 N HSQC/HMQC NMR spectroscopy. Metabolic labeling with multiple fluorinated amino acids provides useful probes for further studying ligand binding and has led to new insight for allosterically regulating transcription-factor protein interactions with small-molecule ligands.
Collapse
Affiliation(s)
- Clifford T Gee
- Department of Chemistry, University of Minnesota, 207 Pleasant Street, SE, Minneapolis, MN, 55455, USA
| | - Keith E Arntson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street, SE, Minneapolis, MN, 55455, USA
| | - Edward J Koleski
- Department of Chemistry, University of Minnesota, 207 Pleasant Street, SE, Minneapolis, MN, 55455, USA
| | - Rachel Lynn Staebell
- Department of Chemistry, University of Minnesota, 207 Pleasant Street, SE, Minneapolis, MN, 55455, USA
| | - William C K Pomerantz
- Department of Chemistry, University of Minnesota, 207 Pleasant Street, SE, Minneapolis, MN, 55455, USA
| |
Collapse
|
193
|
Mitra S, Dash R. Structural dynamics and quantum mechanical aspects of shikonin derivatives as CREBBP bromodomain inhibitors. J Mol Graph Model 2018; 83:42-52. [PMID: 29758466 DOI: 10.1016/j.jmgm.2018.04.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/16/2018] [Accepted: 04/23/2018] [Indexed: 10/17/2022]
Abstract
The Proteins involved in the chemical modification of lysine residues in histone, is currently being excessively focused as the therapeutic target for the treatment of cell related diseases like cancer. Among these proteins, the epigenetic reader, CREB-binding protein (CREBBP) bromodomain is one of the most prominent targets for effective anticancer drug design, which is responsible for the reorganization of acetylated histone lysine residues. Therefore, this study employed an integrative approach of structure based drug design, in combination with Molecular Dynamics (MD) and QM/MM study to identify as well as to describe the binding mechanism of two shikonin derivatives, acetylshikonin and propionylshikonin as inhibitors of CREBBP bromodomain. Here induced fit docking strategy was employed to explore the important intrinsic interactions of ligands with CREBBP bromodomain, consistently molecular dynamics simulation with two different methods and binding energy calculations by MM-GBSA and MM-PBSA were adopted to determine the stability of intermolecular interactions between protein and ligands. The results showed that both these derivatives made direct contacts with the important conserved residues of the active site, where propionylshikonin demonstrated stronger binding and stability than acetylshikonin, according to molecular dynamics simulation and binding free energy calculations. Further, QM/MM energy calculation was employed to study the chemical reactivity of the propionylshikonin and also to describe the mechanism of non bonded interactions between the propionylshikonin and CREBBP bromodomain. Though this study demands in vitro and in vivo experiments to evaluate the efficiency of the compound, these insights would assist to design more potent CREBBP bromodomain inhibitor, guiding the site of modification of propionylshikonin moiety for designing selective inhibitors.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Pharmacy, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Raju Dash
- Molecular Modeling & Drug Design Laboratory (MMDDL), Pharmacology Research Division, Bangladesh Council of Scientific & Industrial Research (BCSIR), Chittagong, 4220, Bangladesh; Department of Biochemistry and Biotechnology, University of Science & Technology Chittagong, Chittagong, 4202, Bangladesh.
| |
Collapse
|
194
|
Li ZJ, Sui XL, Yang XB, Sun W. Similarity of regulatory network between leukemia stem cells and normal hemopoietic stem cells. INFECTION INTERNATIONAL 2018. [DOI: 10.1515/ii-2017-0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractTo reveal the biology of AML, we compared gene-expression profiles between normal hematopoietic cells from 38 healthy donors and leukemic blasts (LBs) from 26 AML patients. We defined the comparison of LB and unselected BM as experiment 1, LB and CD34+ isolated from BM as experiment 2, LB and unselected PB as experiment 3, and LB and CD34+ isolated from PB as experiment 4. Then, protein–protein interaction network of DEGs was constructed to identify critical genes. Regulatory impact factors were used to identify critical transcription factors from the differential co-expression network constructed via reanalyzing the microarray profile from the perspective of differential co-expression. Gene ontology enrichment was performed to extract biological meaning. The comparison among the number of DEGs obtained in four experiments showed that cells did not tend to differentiation and CD34+ was more similar to cancer stem cells. Based on the results of protein–protein interaction network,CREBBP,F2RL1,MCM2, andTP53were respectively the key genes in experiments 1, 2, 3, and 4. From gene ontology analysis, we found that immune response was the most common one in four stages. Our results might provide a platform for determining the pathology and therapy of AML.
Collapse
|
195
|
Berlow RB, Dyson HJ, Wright PE. Expanding the Paradigm: Intrinsically Disordered Proteins and Allosteric Regulation. J Mol Biol 2018; 430:2309-2320. [PMID: 29634920 DOI: 10.1016/j.jmb.2018.04.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 11/30/2022]
Abstract
Allosteric regulatory processes are implicated at all levels of biological function. Recent advances in our understanding of the diverse and functionally significant class of intrinsically disordered proteins have identified a multitude of ways in which disordered proteins function within the confines of the allosteric paradigm. Allostery within or mediated by intrinsically disordered proteins ensures robust and efficient signal integration through mechanisms that would be extremely unfavorable or even impossible for globular protein interaction partners. Here, we highlight recent examples that indicate the breadth of biological outcomes that can be achieved through allosteric regulation by intrinsically disordered proteins. Ongoing and future work in this rapidly evolving area of research will expand our appreciation of the central role of intrinsically disordered proteins in ensuring the fidelity and efficiency of cellular regulation.
Collapse
Affiliation(s)
- Rebecca B Berlow
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - H Jane Dyson
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Peter E Wright
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
196
|
Dutto I, Scalera C, Prosperi E. CREBBP and p300 lysine acetyl transferases in the DNA damage response. Cell Mol Life Sci 2018; 75:1325-1338. [PMID: 29170789 PMCID: PMC11105205 DOI: 10.1007/s00018-017-2717-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
The CREB-binding protein (CREBBP, or in short CBP) and p300 are lysine (K) acetyl transferases (KAT) belonging to the KAT3 family of proteins known to modify histones, as well as non-histone proteins, thereby regulating chromatin accessibility and transcription. Previous studies have indicated a tumor suppressor function for these enzymes. Recently, they have been found to acetylate key factors involved in DNA replication, and in different DNA repair processes, such as base excision repair, nucleotide excision repair, and non-homologous end joining. The growing list of CBP/p300 substrates now includes factors involved in DNA damage signaling, and in other pathways of the DNA damage response (DDR). This review will focus on the role of CBP and p300 in the acetylation of DDR proteins, and will discuss how this post-translational modification influences their functions at different levels, including catalytic activity, DNA binding, nuclear localization, and protein turnover. In addition, we will exemplify how these functions may be necessary to efficiently coordinate the spatio-temporal response to DNA damage. CBP and p300 may contribute to genome stability by fine-tuning the functions of DNA damage signaling and DNA repair factors, thereby expanding their role as tumor suppressors.
Collapse
Affiliation(s)
- Ilaria Dutto
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy
- IRB, Carrer Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Claudia Scalera
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Ennio Prosperi
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
197
|
Daudt C, Da Silva FRC, Lunardi M, Alves CBDT, Weber MN, Cibulski SP, Alfieri AF, Alfieri AA, Canal CW. Papillomaviruses in ruminants: An update. Transbound Emerg Dis 2018; 65:1381-1395. [DOI: 10.1111/tbed.12868] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Indexed: 02/06/2023]
Affiliation(s)
- C. Daudt
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
- Centro de Ciências Biológicas e da Natureza; Universidade Federal do Acre; Rio Branco AC Brazil
| | - F. R. C. Da Silva
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
- Centro de Ciências Biológicas e da Natureza; Universidade Federal do Acre; Rio Branco AC Brazil
| | - M. Lunardi
- Laboratório de Virologia Animal; Departamento de Medicina Veterinária Preventiva; Universidade Estadual de Londrina; Londrina PR Brazil
- Laboratório de Microbiologia Veterinária; Hospital Escola Veterinário; Universidade de Cuiabá; Várzea Grande MT Brazil
| | - C. B. D. T. Alves
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
| | - M. N. Weber
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
| | - S. P. Cibulski
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
| | - A. F. Alfieri
- Laboratório de Virologia Animal; Departamento de Medicina Veterinária Preventiva; Universidade Estadual de Londrina; Londrina PR Brazil
| | - A. A. Alfieri
- Laboratório de Virologia Animal; Departamento de Medicina Veterinária Preventiva; Universidade Estadual de Londrina; Londrina PR Brazil
- Laboratório Multiusuário em Saúde Animal; Unidade de Biologia Molecular; Universidade Estadual de Londrina; Londrina PR Brazil
| | - C. W. Canal
- Laboratório de Virologia Veterinária; Faculdade de Veterinária; Universidade Federal do Rio Grande do Sul; Porto Alegre RS Brazil
| |
Collapse
|
198
|
Koli S, Mukherjee A, Reddy KVR. Retinoic acid triggers c-kit gene expression in spermatogonial stem cells through an enhanceosome constituted between transcription factor binding sites for retinoic acid response element (RARE), spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1) and E26 transformation-specific (ETS). Reprod Fertil Dev 2018; 29:521-543. [PMID: 28442062 DOI: 10.1071/rd15145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/14/2015] [Indexed: 12/19/2022] Open
Abstract
Restricted availability of retinoic acid (RA) in the testicular milieu regulates transcriptional activity of c-kit (KIT, CD117), which aids in the determination of spermatogonial stem-cell differentiation. The effect of RA on c-kit has been reported previously, but its mode of genomic action remains unresolved. We studied the molecular machinery guiding RA responsiveness to the c-kit gene using spermatogonial stem-cell line C18-4 and primary spermatogonial cells. A novel retinoic acid response element (RARE) positioned at -989 nucleotides upstream of the transcription start site (TSS) was identified, providing a binding site for a dimeric RA receptor (i.e. retinoic acid receptor gamma (RARγ) and retinoic X receptor). RA treatment influenced c-kit promoter activity, along with endogenous c-kit expression in C18-4 cells. A comprehensive promoter deletion assay using the pGL3B reporter system characterised the region spanning -271bp and -1011bp upstream of the TSS, which function as minimal promoter and maximal promoter, respectively. In silico analysis predicted that the region -1011 to +58bp comprised the distal enhancer RARE and activators such as spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1), specificity protein 1 (SP1) and four E26 transformation-specific (ETS) tandem binding sites at the proximal region. Gel retardation and chromatin immunoprecipitation (ChIP) assays showed binding for RARγ, PU.1 and SP1 to the predicted consensus binding sequences, whereas GABPα occupied only two out of four ETS binding sites within the c-kit promoter region. We propose that for RA response, an enhanceosome is orchestrated through scaffolding of a CREB-binding protein (CBP)/p300 molecule between RARE and elements in the proximal promoter region, controlling germ-line expression of the c-kit gene. This study outlines the fundamental role played by RARγ, along with other non-RAR transcription factors (PU.1, SP1 and GABPα), in the regulation of c-kit expression in spermatogonial stem cells in response to RA.
Collapse
Affiliation(s)
- Swanand Koli
- Division of Molecular Immunology and Microbiology, National Institute for Research in Reproductive Health, Indian Council of Medical Research, J.M Street, Parel, Mumbai-400 012, India
| | - Ayan Mukherjee
- Department of Biological Science, Kent State University, Kent, OH 44240, USA
| | - Kudumula Venkata Rami Reddy
- Division of Molecular Immunology and Microbiology, National Institute for Research in Reproductive Health, Indian Council of Medical Research, J.M Street, Parel, Mumbai-400 012, India
| |
Collapse
|
199
|
Cottone E, Orso F, Biglia N, Sismondi P, De Bortoli M. Role of Coactivators and Corepressors in Steroid and Nuclear Receptor Signaling: Potential Markers of Tumor Growth and Drug Sensitivity. Int J Biol Markers 2018; 16:151-66. [PMID: 11605727 DOI: 10.1177/172460080101600301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nuclear receptors regulate target gene expression in response to steroid and thyroid hormones, retinoids, vitamin D and other ligands. These ligand-dependent transcription factors function by contacting various nuclear cooperating proteins, called coactivators and corepressors, which mediate local chromatin remodeling as well as communication with the basal transcriptional apparatus. Nuclear receptors and their coregulatory proteins play a role in cancer and other diseases, one leading example being the estrogen receptor pathway in breast cancer. Coregulators are often present in limiting amounts in cell nuclei and modifications of their level of expression and/or structure lead to alterations in nuclear receptor functioning, which may be as pronounced as a complete inversion of signaling, i.e. from stimulating to repressing certain genes in response to an identical stimulus. In addition, hemizygous knock-out of certain coactivator genes has been demonstrated to produce cancer-prone phenotypes in mice. Thus, assessment of coactivator and corepressor expression and structure in tumors may turn out to be essential to determine the role of nuclear receptors in cancer and to predict prognosis and response to therapy.
Collapse
Affiliation(s)
- E Cottone
- Department of Animal and Human Biology, University of Turin, Italy
| | | | | | | | | |
Collapse
|
200
|
In silico quest of selective naphthyl-based CREBBP bromodomain inhibitor. In Silico Pharmacol 2018; 6:1. [PMID: 30607314 DOI: 10.1007/s40203-018-0038-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 03/02/2018] [Indexed: 10/17/2022] Open
Abstract
The reader proteins like bromodomains have recently gained increased attentions in the area of epigenetic drug discovery, as they are the potent regulators in gene transcription process. Among the other bromodomains, cAMP response element-binding protein (CREB) binding protein or CREBBP bomodomain involved in various cancer progressions and therefore, efforts to develop specific inhibitors of CREBBP bomodomain are of clinical value. In this study, we tried to identify selective CREBBP bromodomain inhibitor, which was accomplished by using molecular docking, free energy calculation and molecular dynamics (MD) simulation studies, considering a series of naphthyl based compounds. The docking procedure was validated by comparing root mean square deviations (RMSDs) of crystallographic complex to docked complex. Favorable electrostatic interactions with the Arg1173 side chain were considered to attain selectivity for CREBBP bromodomain over other human bromodomain subfamilies. We found that naphthyl-based compounds have greater binding affinities towards the CREBBP bromodomain, and formed non-bonded interactions with various side chain residues that are important for bromodomain inhibition. From detailed investigation by induced fit docking, compound 31 was found to have favorable electrostatic interactions with the Arg1173 side chain by forming conventional hydrogen bonds. This result was further confirmed by analyzing hydrogen bond occupancy and bonding distance during the molecular dynamics simulation. We believe that these findings offer useful insight for the designing of target specific new bromodomain inhibitor and also promote further structure guided synthesis of analogues for identification of potent CREBBP bromodomain inhibitors as well as detailed in vitro and in vivo analyses.
Collapse
|