1
|
Neves S, Pacheco S, Vaz F, James P, Simões T, Penque D. Occupational second-hand smoke exposure: A comparative shotgun proteomics study on nasal epithelia from healthy restaurant workers. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104459. [PMID: 38685369 DOI: 10.1016/j.etap.2024.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Non-smokers exposed to second-hand smoke (SHS) present risk of developing tobacco smoke-associated pathologies. To investigate the airway molecular response to SHS exposure that could be used in health risk assessment, comparative shotgun proteomics was performed on nasal epithelium from a group of healthy restaurant workers, non-smokers (never and former) exposed and not exposed to SHS in the workplace. HIF1α-glycolytic targets (GAPDH, TPI) and proteins related to xenobiotic metabolism, cell proliferation and differentiation leading to cancer (ADH1C, TUBB4B, EEF2) showed significant modulation in non-smokers exposed. In never smokers exposed, enrichment of glutathione metabolism pathway and EEF2-regulating protein synthesis in genotoxic response were increased, while in former smokers exposed, proteins (LYZ, ATP1A1, SERPINB3) associated with tissue damage/regeneration, apoptosis inhibition and inflammation that may lead to asthma, COPD or cancer, were upregulated. The identified proteins are potential response and susceptibility/risk biomarkers for SHS exposure.
Collapse
Affiliation(s)
- Sofia Neves
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal.
| | - Solange Pacheco
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal
| | - Fátima Vaz
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal
| | - Peter James
- Protein Technology Laboratory, Department of Immunotechnology, Lund University, Sweden
| | - Tânia Simões
- CECAD Cologne-Excellence in Aging Research University of Cologne, Germany
| | - Deborah Penque
- Laboratory of Proteomics, Human Genetics Department, National Institute of Health Dr. Ricardo Jorge, INSA I.P, Lisbon, Portugal; Center for Toxicogenomics and Human Health, ToxOmics, NOVA Medical School-FCM, UNL, Lisbon, Portugal
| |
Collapse
|
2
|
Nabekura T, Deborah EA, Tahara S, Arai Y, Love PE, Kako K, Fukamizu A, Muratani M, Shibuya A. Themis2 regulates natural killer cell memory function and formation. Nat Commun 2023; 14:7200. [PMID: 37938555 PMCID: PMC10632368 DOI: 10.1038/s41467-023-42578-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023] Open
Abstract
Immunological memory is a hallmark of the adaptive immune system. Although natural killer (NK) cells are innate immune cells important for the immediate host defence, they can differentiate into memory NK cells. The molecular mechanisms controlling this differentiation are yet to be fully elucidated. Here we identify the scaffold protein Themis2 as a critical regulator of memory NK cell differentiation and function. Themis2-deficient NK cells expressing Ly49H, an activating NK receptor for the mouse cytomegalovirus (MCMV) antigen m157, show enhanced differentiation into memory NK cells and augment host protection against MCMV infection. Themis2 inhibits the effector function of NK cells after stimulation of Ly49H and multiple activating NK receptors, though not specific to memory NK cells. Mechanistically, Themis2 suppresses Ly49H signalling by attenuating ZAP70/Syk phosphorylation, and it also translocates to the nucleus, where it promotes Zfp740-mediated repression to regulate the persistence of memory NK cells. Zfp740 deficiency increases the number of memory NK cells and enhances the effector function of memory NK cells, which further supports the relevance of the Themis2-Zfp740 pathway. In conclusion, our study shows that Themis2 quantitatively and qualitatively regulates NK cell memory formation.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan.
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, 305-8575, Japan.
| | - Elfira Amalia Deborah
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Saeko Tahara
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Yuya Arai
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Koichiro Kako
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Shibuya
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan.
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
3
|
Vazquez-Sandoval A, Velez-delValle C, Hernández-Mosqueira C, Marsch-Moreno M, Ayala-Sumuano JT, Kuri-Harcuch W. FAM129B is a cooperative protein that regulates adipogenesis. Biochem Biophys Res Commun 2023; 638:66-75. [PMID: 36442234 DOI: 10.1016/j.bbrc.2022.11.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
FAM129B is one of Niban-like proteins described in neoplastic cells and implicated in melanoma cell invasion, but no reports have been published on FAM129B and cell differentiation. We show that FAM129B is early and transiently expressed and crucial for 3T3-F442A adipogenesis. Fam129b is expressed downstream of the early genes Cebpb, Klf4, Klf5 and Srebf1a, but upstream of Pparg2 since knockdown of Fam129b blocked Pparg2 expression and adipose differentiation. Glycogen synthase kinase 3 beta activity, a crucial kinase for adipogenesis, and the ERK1/2 are involved in FAM129B phosphorylation as part of the adipogenic program. Phosphorylated FAM129B is crucial for Pparg2 expression and the lipogenic gene expression downstream of Pparg2, and hence for adipogenesis. Fam129b knockdown reduced adipocyte cluster formation and size, regulating commitment and clonal amplification. In vivo, BAT, inguinal and epidydimal fat expressed Fam129b, suggesting a role in adipose tissue development. We conclude that FAM129B is a cooperative protein that regulates differentiation during the early stages of adipogenesis.
Collapse
Affiliation(s)
- Alfredo Vazquez-Sandoval
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Cristina Velez-delValle
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Claudia Hernández-Mosqueira
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Meytha Marsch-Moreno
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico
| | - Jorge-Tonatiuh Ayala-Sumuano
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico; Department of Biomedical Research, IDIX Biotech, Avenida de Los Portones 1151, Queretaro, CP 76100, Mexico
| | - Walid Kuri-Harcuch
- Department of Cell Biology, Center of Research and Advanced Studies (CINVESTAV), IPN Avenida Instituto Politécnico Nacional 2508, Mexico City, CP 07360, Mexico.
| |
Collapse
|
4
|
Discerning asthma endotypes through comorbidity mapping. Nat Commun 2022; 13:6712. [PMID: 36344522 PMCID: PMC9640644 DOI: 10.1038/s41467-022-33628-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Asthma is a heterogeneous, complex syndrome, and identifying asthma endotypes has been challenging. We hypothesize that distinct endotypes of asthma arise in disparate genetic variation and life-time environmental exposure backgrounds, and that disease comorbidity patterns serve as a surrogate for such genetic and exposure variations. Here, we computationally discover 22 distinct comorbid disease patterns among individuals with asthma (asthma comorbidity subgroups) using diagnosis records for >151 M US residents, and re-identify 11 of the 22 subgroups in the much smaller UK Biobank. GWASs to discern asthma risk loci for individuals within each subgroup and in all subgroups combined reveal 109 independent risk loci, of which 52 are replicated in multi-ancestry meta-analysis across different ethnicity subsamples in UK Biobank, US BioVU, and BioBank Japan. Fourteen loci confer asthma risk in multiple subgroups and in all subgroups combined. Importantly, another six loci confer asthma risk in only one subgroup. The strength of association between asthma and each of 44 health-related phenotypes also varies dramatically across subgroups. This work reveals subpopulations of asthma patients distinguished by comorbidity patterns, asthma risk loci, gene expression, and health-related phenotypes, and so reveals different asthma endotypes.
Collapse
|
5
|
da Silva FA, Rodrigues-Ribeiro L, Melo-Braga MN, Passos-Silva DG, Sampaio WO, Gorshkov V, Kjeldsen F, Verano-Braga T, Santos RAS. Phosphoproteomic studies of alamandine signaling in CHO-MrgD and human pancreatic carcinoma cells: An antiproliferative effect is unveiled. Proteomics 2022; 22:e2100255. [PMID: 35652611 DOI: 10.1002/pmic.202100255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022]
Abstract
Alamandine is a heptapeptide from the renin-angiotensin system (RAS) with similar structure/function to angiotensin-(1-7) [ang-(1-7)], but they act via different receptors. It remains elusive whether alamandine is an antiproliferative agent like ang-(1-7). The goal of this study was to evaluate the potential antiproliferative activity of alamandine and the underlying cellular signaling. We evaluated alamandine effect in the tumoral cell lines Mia PaCa-2 and A549, and in the nontumoral cell lines HaCaT, CHO and CHO transfected with the alamandine receptor MrgD (CHO-MrgD). Alamandine was able to reduce the proliferation of the tumoral cell lines in a MrgD-dependent fashion. We did not observe any effect in the nontumoral cell lines tested. We also performed proteomics and phosphoproteomics to study the alamandine signaling in Mia PaCa-2 and CHO-MrgD. Data suggest that alamandine induces a shift from anaerobic to aerobic metabolism in the tumoral cells, induces a negative regulation of PI3K/AKT/mTOR pathway and activates the transcriptional factor FoxO1; events that could explain, at least partially, the observed antiproliferative effect of alamandine. This study provides for the first time a comprehensive investigation of the alamandine signaling in tumoral (Mia PaCa-2) and nontumoral (CHO-MrgD) cells, highlighting the antiproliferative activity of alamandine/MrgD and its possible antitumoral effect.
Collapse
Affiliation(s)
- Filipe Alex da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
| | - Lucas Rodrigues-Ribeiro
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcella Nunes Melo-Braga
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Danielle Gomes Passos-Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
| | - Walkyria Oliveira Sampaio
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
| | - Vladimir Gorshkov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Syddanmark, Denmark
| | - Frank Kjeldsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Syddanmark, Denmark
| | - Thiago Verano-Braga
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
6
|
Zeng G, Lian C, Li W, An H, Han Y, Fang D, Zheng Q. Upregulation of FAM129B protects cardiomyocytes from hypoxia/reoxygenation-induced injury by inhibiting apoptosis, oxidative stress, and inflammatory response via enhancing Nrf2/ARE activation. ENVIRONMENTAL TOXICOLOGY 2022; 37:1018-1031. [PMID: 34995000 DOI: 10.1002/tox.23461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 12/02/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
Family with sequence similarity 129, member B (FAM129B) has been identified as a novel cytoprotective protein that facilitates the survival of detrimentally stimulated cells. However, whether FAM129B is involved in regulating cardiomyocyte survival after myocardial ischemia-reperfusion injury is unknown. The goal of this work was to evaluate the potential role of FAM129B in regulating hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury in vitro. We demonstrated that exposure to H/R markedly downregulated the expression of FAM129B in cardiomyocytes. Functional experiments revealed that the upregulation of FAM129B improved H/R-exposed cardiomyocyte viability, and ameliorated H/R-induced cardiomyocyte apoptosis, the generation of reactive oxygen species (ROS), and pro-inflammatory cytokine release. The upregulation of FAM129B significantly increased the nuclear expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), and reinforced Nrf2/antioxidant response element (ARE) activation in H/R-exposed cardiomyocytes. Moreover, FAM129B modulates Nrf2/ARE signaling in a Kelchlike ECH-associated protein 1-dependent manner. Notably, the inhibition of Nrf2 significantly blocked FAM129B-overexpression-induced cardioprotective effects in H/R-exposed cardiomyocytes. In summary, the findings of our work demonstrate that the upregulation of FAM129B ameliorates H/R-induced cardiomyocyte injury via enhancing Nrf2/ARE activation. Thus, our study indicates that FAM129B may play a role in myocardial ischemia-reperfusion injury and has the potential to be used as a cardioprotective target.
Collapse
Affiliation(s)
- Guangwei Zeng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Cheng Lian
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Wei Li
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Huixian An
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Yang Han
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Dong Fang
- Section 2, Department of Cardiology, Xi'an International Medical Center Hospital, Shaanxi, China
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| |
Collapse
|
7
|
Diana P, Carvalheira GMG. NIBAN1, Exploring its Roles in Cell Survival Under Stress Context. Front Cell Dev Biol 2022; 10:867003. [PMID: 35517496 PMCID: PMC9062034 DOI: 10.3389/fcell.2022.867003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cell survival must quickly activate specific mechanisms that enable to detect changes in the cellular microenvironment. The impact of these cell alteration has direct consequences on cellular homeostasis. Cellular stress, as well as its regulation and implication, has been studied in different pathologies. In this sense, the alteration in NIBAN1 expression seems to act in response to different cellular disturbances. Over the years, the knowledge of NIBAN1 functions has improved, demonstrating its important cell roles, favoring the cell survival under stress context. In response to the disturbances, NIBAN1 seems to be involved in the decision-making process between cell survival and death. The increase in NIBAN1 expression has been related to cellular mechanisms that seek to minimize the damage caused to cellular homeostasis. In this review, the main biological insights attributed to the NIBAN1 gene in different cellular contexts and its role as a mediator of cellular stress are discussed.
Collapse
|
8
|
Dolinski AC, Homola JJ, Jankowski MD, Robinson JD, Owen JC. Differential gene expression reveals host factors for viral shedding variation in mallards ( Anas platyrhynchos) infected with low-pathogenic avian influenza virus. J Gen Virol 2022; 103:10.1099/jgv.0.001724. [PMID: 35353676 PMCID: PMC10519146 DOI: 10.1099/jgv.0.001724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intraspecific variation in pathogen shedding impacts disease transmission dynamics; therefore, understanding the host factors associated with individual variation in pathogen shedding is key to controlling and preventing outbreaks. In this study, ileum and bursa of Fabricius tissues of wild-bred mallards (Anas platyrhynchos) infected with low-pathogenic avian influenza (LPAIV) were evaluated at various post-infection time points to determine genetic host factors associated with intraspecific variation in viral shedding. By analysing transcriptome sequencing data (RNA-seq), we found that LPAIV-infected wild-bred mallards do not exhibit differential gene expression compared to uninfected birds, but that gene expression was associated with cloacal viral shedding quantity early in the infection. In both tissues, immune gene expression was higher in high/moderate shedding birds compared to low shedding birds, and significant positive relationships with viral shedding were observed. In the ileum, expression for host genes involved in viral cell entry was lower in low shedders compared to moderate shedders at 1 day post-infection (DPI), and expression for host genes promoting viral replication was higher in high shedders compared to low shedders at 2 DPI. Our findings indicate that viral shedding is a key factor for gene expression differences in LPAIV-infected wild-bred mallards, and the genes identified in this study could be important for understanding the molecular mechanisms driving intraspecific variation in pathogen shedding.
Collapse
Affiliation(s)
- Amanda C. Dolinski
- Department of Fisheries and Wildlife, Michigan State
University, East Lansing, MI
| | - Jared J. Homola
- Department of Fisheries and Wildlife, Michigan State
University, East Lansing, MI
| | - Mark D. Jankowski
- Department of Fisheries and Wildlife, Michigan State
University, East Lansing, MI
- U.S. Environmental Protection Agency, Region 10, Seattle,
WA 98101
| | - John D. Robinson
- Department of Fisheries and Wildlife, Michigan State
University, East Lansing, MI
| | - Jennifer C. Owen
- Department of Fisheries and Wildlife, Michigan State
University, East Lansing, MI
- Department of Large Animal Clinical Sciences, Michigan
State University, East Lansing, MI, USA
| |
Collapse
|
9
|
White BS, Khan SA, Mason MJ, Ammad-Ud-Din M, Potdar S, Malani D, Kuusanmäki H, Druker BJ, Heckman C, Kallioniemi O, Kurtz SE, Porkka K, Tognon CE, Tyner JW, Aittokallio T, Wennerberg K, Guinney J. Bayesian multi-source regression and monocyte-associated gene expression predict BCL-2 inhibitor resistance in acute myeloid leukemia. NPJ Precis Oncol 2021; 5:71. [PMID: 34302041 PMCID: PMC8302655 DOI: 10.1038/s41698-021-00209-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 06/22/2021] [Indexed: 11/09/2022] Open
Abstract
The FDA recently approved eight targeted therapies for acute myeloid leukemia (AML), including the BCL-2 inhibitor venetoclax. Maximizing efficacy of these treatments requires refining patient selection. To this end, we analyzed two recent AML studies profiling the gene expression and ex vivo drug response of primary patient samples. We find that ex vivo samples often exhibit a general sensitivity to (any) drug exposure, independent of drug target. We observe that this "general response across drugs" (GRD) is associated with FLT3-ITD mutations, clinical response to standard induction chemotherapy, and overall survival. Further, incorporating GRD into expression-based regression models trained on one of the studies improved their performance in predicting ex vivo response in the second study, thus signifying its relevance to precision oncology efforts. We find that venetoclax response is independent of GRD but instead show that it is linked to expression of monocyte-associated genes by developing and applying a multi-source Bayesian regression approach. The method shares information across studies to robustly identify biomarkers of drug response and is broadly applicable in integrative analyses.
Collapse
Affiliation(s)
- Brian S White
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA.
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
| | - Suleiman A Khan
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Mike J Mason
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
| | - Muhammad Ammad-Ud-Din
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Disha Malani
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Heikki Kuusanmäki
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Brian J Druker
- Howard Hughes Medical Institute, Portland, OR, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Caroline Heckman
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Scilifelab, Karolinska Institute, Solna, Sweden
| | - Stephen E Kurtz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kimmo Porkka
- HUS Comprehensive Cancer Center, Hematology Research Unit Helsinki and iCAN Digital Precision Cancer Center Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Cristina E Tognon
- Howard Hughes Medical Institute, Portland, OR, USA
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W Tyner
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Justin Guinney
- Computational Oncology, Sage Bionetworks, Seattle, WA, USA
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| |
Collapse
|
10
|
Schmidlin CJ, Tian W, Dodson M, Chapman E, Zhang DD. FAM129B-dependent activation of NRF2 promotes an invasive phenotype in BRAF mutant melanoma cells. Mol Carcinog 2021; 60:331-341. [PMID: 33684228 DOI: 10.1002/mc.23295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022]
Abstract
Incidence of melanoma continues to rise in the United States with ~100,000 new cases diagnosed in 2019. While the 5-year survival rate of melanoma is 99% when localized, the rate of survival drops to 22.5% when distant disease is detected. As such, an area of great interest is understanding the mechanisms that promote melanoma metastasis so that better potential therapeutic targets can be discovered. Herein, we demonstrate that activation of NRF2 by FAM129B contributes to increased metastatic potential of BRAF V600E mutant melanoma cells. Specifically, FAM129B induces NRF2 by competing for Kelch-like ECH-associated protein 1 (KEAP1) binding (the negative regulator of NRF2) via an ETGE motif. Furthermore, we show that phosphorylation of FAM129B plays a role in mediating the interaction between FAM129B and KEAP1, as the phosphorylation status of FAM129B dictates its subcellular localization. When phosphorylated, FAM129B is found primarily in the cytosol where it can bind to KEAP1, but upon inhibition of mitogen-activated protein kinase kinase activity, FAM129B is localized to the cell membrane and no longer interacts with KEAP1. In BRAF V600E mutant melanoma, the mitogen-activated protein kinase pathway leads to hyperphosphorylation of FAM129B, and therefore FAM129B localizes to the cytosol, binds KEAP1, and upregulates NRF2. Importantly, genetic modulation or pharmacological inhibition that results in a decrease in FAM129B protein level or its phosphorylation decreases migration and invasion of mutant melanoma in an NRF2-dependent manner. Overall, these data indicate that phosphorylation of FAM129B plays a significant role in driving the metastatic potential of BRAF V600E melanoma via upregulation of the NRF2 signaling pathway.
Collapse
Affiliation(s)
- Cody J Schmidlin
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Wang Tian
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Matthew Dodson
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Eli Chapman
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Donna D Zhang
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA.,Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
11
|
Lee D. miR-769-5p is associated with prostate cancer recurrence and modulates proliferation and apoptosis of cancer cells. Exp Ther Med 2021; 21:335. [PMID: 33732308 PMCID: PMC7903391 DOI: 10.3892/etm.2021.9766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRs) are relevant in biological processes, including human prostate cancer. In the present study, the role of miR-769-5p and its targets in prostate cancer were explored. Publicly available data on expression of genes, miRs and disease-free survival of patients with prostate cancer were analyzed along with RNAseq of transfected cell lines. miR-769-5p expression was inversely associated with patient survival and in vitro assays indicated that its inhibition reduced the proliferation and increased apoptosis of prostate cancer cells. miR-769-5p was revealed to target Rho GTPase activating protein 10 (ARHGAP10) and increased expression of ARHGAP10 in tumors was determined to be associated with a favorable prognosis regarding disease-free survival. Of note, ARHGAP10 is a purported tumor suppressor in ovarian cancer, where it inhibits cell division cycle 42 (CDC42) activity and increases apoptosis. Similar effects were observed in prostate cancer cells, where miR-769-5p inhibition increased ARHGAP10 and led to reduced CDC42 activity. Furthermore, miR-769-5p inhibition increased apoptosis, which was partly reversed by additional knockdown of ARHGAP10. These results suggested that miR-769-5p is an oncogene targeting ARHGAP10, which in turn is a candidate tumor suppressor in prostate cancer.
Collapse
Affiliation(s)
- Daniel Lee
- Medical Oncology Service and The Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Seo YA, Choi EK, Aring L, Paschall M, Iwase S. Transcriptome Analysis of the Cerebellum of Mice Fed a Manganese-Deficient Diet. Front Genet 2020; 11:558725. [PMID: 33408735 PMCID: PMC7780674 DOI: 10.3389/fgene.2020.558725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/13/2020] [Indexed: 11/13/2022] Open
Abstract
Manganese (Mn), primarily acquired through diet, is required for brain function and development. Epidemiological studies have found an association between both low and high levels of Mn and impaired neurodevelopment in children. Recent genetic studies have revealed that patients with congenital Mn deficiency display severe psychomotor disability and cerebral and cerebellar atrophy. Although the impact of Mn on gene expression is beginning to be appreciated, Mn-dependent gene expression remains to be explored in vertebrate animals. The goal of this study was to use a mouse model to define the impact of a low-Mn diet on brain metal levels and gene expression. We interrogated gene expression changes in the Mn-deficient mouse brain at the genome-wide scale by RNA-seq analysis of the cerebellum of mice fed low or normal Mn diets. A total of 137 genes were differentially expressed in Mn-deficient cerebellums compared with Mn-adequate cerebellums (Padj < 0.05). Mn-deficient mice displayed downregulation of key pathways involved with "focal adhesion," "neuroactive ligand-receptor interaction," and "cytokine-cytokine receptor interaction" and upregulation of "herpes simplex virus 1 infection," "spliceosome," and "FoxO signaling pathway." Reactome pathway analysis identified upregulation of the splicing-related pathways and transcription-related pathways, as well as downregulation of "metabolism of carbohydrate," and "extracellular matrix organization," and "fatty acid metabolism" reactomes. The recurrent identifications of splicing-related pathways suggest that Mn deficiency leads to upregulation of splicing machineries and downregulation of diverse biological pathways.
Collapse
Affiliation(s)
- Young Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Eun-Kyung Choi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Luisa Aring
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Molly Paschall
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Shigeki Iwase
- Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
13
|
Qian K, Xu JX, Deng Y, Peng H, Peng J, Ou CM, Liu Z, Jiang LH, Tai YH. Signaling pathways of genetic variants and miRNAs in the pathogenesis of myasthenia gravis. Gland Surg 2020; 9:1933-1944. [PMID: 33447544 PMCID: PMC7804555 DOI: 10.21037/gs-20-39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Myasthenia gravis (MG) is a chronic autoimmune neuromuscular disorder causing muscle weakness and characterized by a defect in synaptic transmission at the neuromuscular junction. The pathogenesis of this disease remains unclear. We aimed to predict the key signaling pathways of genetic variants and miRNAs in the pathogenesis of MG, and identify the key genes among them. METHODS We searched published information regarding associated single nucleotide polymorphisms (SNPs) and differentially-expressed miRNAs in MG cases. We search of SNPs and miRNAs in literature databases about MG, then we used bioinformatic tools to predict target genes of miRNAs. Moreover, functional enrichment analysis for key genes was carried out utilizing the Cytoscape-plugin, known as ClueGO. These key genes were mapped to STRING database to construct a protein-protein interaction (PPI) network. Then a miRNA-target gene regulatory network was established to screen key genes. RESULTS Five genes containing SNPs associated with MG risk were involved in the inflammatory bowel disease (IBD) signaling pathway, and FoxP3 was the key gene. MAPK1, SMAD4, SMAD2 and BCL2 were predicted to be targeted by the 18 miRNAs and to act as the key genes in adherens, junctions, apoptosis, or cancer-related pathways respectively. These five key genes containing SNPs or targeted by miRNAs were found to be involved in negative regulation of T cell differentiation. CONCLUSIONS We speculate that SNPs cause the genes to be defective or the miRNAs to downregulate the factors that subsequently negatively regulate regulatory T cells and trigger the onset of MG.
Collapse
Affiliation(s)
- Kai Qian
- Faculty of Life and Biotechnology, Kunming University of Science and Technology, Kunming, China
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jia-Xin Xu
- Department of Cardiovascular surgery, Yan’ an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi Deng
- Department of Oncology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Peng
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jun Peng
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Chun-Mei Ou
- Department of Cardiovascular surgery, Institute of the First People’s Hospital of Yunnan Province, Kunming, China
| | - Zu Liu
- Department of Cardiovascular surgery, Yan’ an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li-Hong Jiang
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yong-Hang Tai
- School of Electronic Information in the Yunnan Normal University, Kunming, China
| |
Collapse
|
14
|
Structural Insight on Functional Regulation of Human MINERVA Protein. Int J Mol Sci 2020; 21:ijms21218186. [PMID: 33142954 PMCID: PMC7663100 DOI: 10.3390/ijms21218186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 11/17/2022] Open
Abstract
MINERVA (melanoma invasion by ERK), also known as FAM129B, is a member of the FAM129 protein family, which is only present in vertebrates. MINERVA is involved in key signaling pathways regulating cell survival, proliferation and apoptosis and found upregulated in many types of cancer promoting invasion. However, the exact function of the protein remains elusive. X-ray crystallographic methods were implemented to determine the crystal structure of MINERVAΔC, lacking C-terminal flexible region. Trypsin digestion was required before crystallization to obtain diffraction-quality crystals. While the N-terminal pleckstrin homology (PH) domain exhibits the typical fold of PH domains, lipid binding assay indicates specific affinity towards phosphatidic acid and inositol 3-phosphate. A helix-rich domain that constitutes the rest of the molecule demonstrates a novel L-shaped fold that encompasses the PH domain. The overall structure of MINERVAΔC with binding assays and cell-based experiments suggest plasma membrane association of MINERVA and its function seem to be tightly regulated by various motifs within the C-terminal flexible region. Elucidation of MINERVAΔC structure presents a novel fold for an α-helix bundle domain that would provide a binding platform for interacting partners.
Collapse
|
15
|
Ryu HS, Park KW, Choi N, Kim J, Park YM, Jo S, Kim MJ, Kim YJ, Kim J, Kim K, Koh SB, Chung SJ. Genomic Analysis Identifies New Loci Associated With Motor Complications in Parkinson's Disease. Front Neurol 2020; 11:570. [PMID: 32733355 PMCID: PMC7358548 DOI: 10.3389/fneur.2020.00570] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Parkinson's disease (PD) is a common neurodegenerative disorder, characterized by a clinical symptomatology involving both motor and non-motor symptoms. Motor complications associated with long-term dopaminergic treatment include motor fluctuations and levodopa-induced dyskinesia (LID), which may have a major impact on the quality of life. The clinical features and onset time of motor complications in the disease course are heterogeneous, and the etiology remains unknown. Objective: We aimed to identify genomic variants associated with the development of motor fluctuations and LID at 5 years after the onset of PD. Methods: Genomic data were obtained using Affymetrix Axiom KORV1.1 array, including an imputation genome-wide association study (GWAS) grid and other GWAS loci; functional variants of the non-synonymous exome; pharmacogenetic variants; variants in genes involved in absorption, distribution, metabolism, and excretion of drugs; and expression quantitative trait loci in 741 patients with PD. Results: FAM129B single-nucleotide polymorphism (SNP) rs10760490 was nominally associated with the occurrence of motor fluctuations at 5 years after the onset of PD [odds ratio (OR) = 2.9, 95% confidence interval (CI) = 1.8-4.8, P = 6.5 × 10-6]. GALNT14 SNP rs144125291 was significantly associated with the occurrence of LID (OR = 5.5, 95% CI = 2.9-10.3, P = 7.88 × 10-9) and was still significant after Bonferroni correction. Several other genetic variants were associated with the occurrence of motor fluctuations or LID, but the associations were not significant after Bonferroni correction. Conclusion: This study identified new loci associated with the occurrence of motor fluctuations and LID at 5 years after the onset of PD. However, further studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Ho-Sung Ryu
- Department of Neurology, Kyungpook National University Hospital, Daegu, South Korea
| | - Kye Won Park
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Nari Choi
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jinhee Kim
- Department of Neurology & Parkinson's Disease Center, Guro Hospital, Korea University, Seoul, South Korea
| | - Young-Min Park
- Department of Neurology, Dobong Hospital, Seoul, South Korea
| | - Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Mi-Jung Kim
- Department of Neurology, Bobath Memorial Hospital, Seongnam-si, South Korea
| | - Young Jin Kim
- Department of Neurology, Best Heals Hospital, Ansan-si, South Korea
| | - Juyeon Kim
- Department of Neurology, Metro Hospital, Anyang, South Korea
| | - Kiju Kim
- Department of Neurology, The Good Light Hospital, Gwangju, South Korea
| | - Seong-Beom Koh
- Department of Neurology & Parkinson's Disease Center, Guro Hospital, Korea University, Seoul, South Korea
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Plum T, Wang X, Rettel M, Krijgsveld J, Feyerabend TB, Rodewald HR. Human Mast Cell Proteome Reveals Unique Lineage, Putative Functions, and Structural Basis for Cell Ablation. Immunity 2020; 52:404-416.e5. [DOI: 10.1016/j.immuni.2020.01.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/13/2019] [Accepted: 01/22/2020] [Indexed: 12/25/2022]
|
17
|
Santos Silva DBD, Fonseca LFS, Magalhães AFB, Muniz MMM, Baldi F, Ferro JA, Chardulo LAL, Pinheiro DG, Albuquerque LGD. Transcriptome profiling of muscle in Nelore cattle phenotypically divergent for the ribeye muscle area. Genomics 2019; 112:1257-1263. [PMID: 31351181 DOI: 10.1016/j.ygeno.2019.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/28/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
This study aimed to use RNA-Seq to identify differentially expressed genes (DEGs) in muscle of uncastrated Nelore males phenotypically divergent for ribeye muscle area (REA). A total of 80 animals were phenotyped for REA, and 15 animals each with the highest REA and the lowest REA were selected for analyses. DEGs found (N = 288) belonging to families related to muscle cell growth, development, motility and proteolysis, such as actin, myosin, collagen, integrin, solute carrier, ubiquitin and kelch-like. Functional analysis showed that many of the significantly enriched gene ontology terms were closely associated with muscle development, growth, and degradation. Through co-expression network analysis, we predicted three hub genes (PPP3R1, FAM129B and UBE2G1), these genes are involved in muscle growth, proteolysis and immune system. The genes expression levels and its biological process found this study may result in differences in muscle deposition, and therefore, Nelore animals with different REA proportions.
Collapse
Affiliation(s)
- Danielly Beraldo Dos Santos Silva
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil; National Council for Scientific and Technological Development (CNPq), Brasilia, DF, Brazil.
| | - Larissa Fernanda Simielli Fonseca
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil; National Council for Scientific and Technological Development (CNPq), Brasilia, DF, Brazil
| | | | | | - Fernando Baldi
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil; National Council for Scientific and Technological Development (CNPq), Brasilia, DF, Brazil
| | - Jesus Aparecido Ferro
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil; National Council for Scientific and Technological Development (CNPq), Brasilia, DF, Brazil
| | | | - Daniel Guariz Pinheiro
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | - Lucia Galvão de Albuquerque
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil; National Council for Scientific and Technological Development (CNPq), Brasilia, DF, Brazil.
| |
Collapse
|
18
|
Cheng KC, Lin RJ, Cheng JY, Wang SH, Yu JC, Wu JC, Liang YJ, Hsu HM, Yu J, Yu AL. FAM129B, an antioxidative protein, reduces chemosensitivity by competing with Nrf2 for Keap1 binding. EBioMedicine 2019; 45:25-38. [PMID: 31262713 PMCID: PMC6642435 DOI: 10.1016/j.ebiom.2019.06.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The transcription factor Nrf2 is a master regulator of antioxidant response. While Nrf2 activation may counter increasing oxidative stress in aging, its activation in cancer can promote cancer progression and metastasis, and confer resistance to chemotherapy and radiotherapy. Thus, Nrf2 has been considered as a key pharmacological target. Unfortunately, there are no specific Nrf2 inhibitors for therapeutic application. Moreover, high Nrf2 activity in many tumors without Keap1 or Nrf2 mutations suggests that alternative mechanisms of Nrf2 regulation exist. METHODS Interaction of FAM129B with Keap1 is demonstrated by immunofluorescence, colocalization, co-immunoprecipitation and mammalian two-hybrid assay. Antioxidative function of FAM129B is analyzed by measuring ROS levels with DCF/flow cytometry, Nrf2 activation using luciferase reporter assay and determination of downstream gene expression by qPCR and wester blotting. Impact of FAM129B on in vivo chemosensitivity is examined in mice bearing breast and colon cancer xenografts. The clinical relevance of FAM129B is assessed by qPCR in breast cancer samples and data mining of publicly available databases. FINDINGS We have demonstrated that FAM129B in cancer promotes Nrf2 activity by reducing its ubiquitination through competition with Nrf2 for Keap1 binding via its DLG and ETGE motifs. In addition, FAM129B reduces chemosensitivity by augmenting Nrf2 antioxidative signaling and confers poor prognosis in breast and lung cancer. INTERPRETATION These findings demonstrate the important role of FAM129B in Nrf2 activation and antioxidative response, and identify FMA129B as a potential therapeutic target. FUND: The Chang Gung Medical Foundation (Taiwan) and the Ministry of Science and Technology (Taiwan).
Collapse
Affiliation(s)
- Kai-Chun Cheng
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Ruey-Jen Lin
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jing-Yan Cheng
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Hung Wang
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Jyh-Cherng Yu
- General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jen-Chine Wu
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Yuh-Jin Liang
- Translational Research Division, Medical Research Department, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Huan-Ming Hsu
- General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - John Yu
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, University of California in San Diego, San Diego, CA, USA.
| |
Collapse
|
19
|
Zhou X, Yang F, Zhang Q, Miao Y, Hu X, Li A, Hou G, Wang Q, Kang J. FAM129B promoted tumor invasion and proliferation via facilitating the phosphorylation of FAK signaling and associated with adverse clinical outcome of non-small cell lung cancer patients. Onco Targets Ther 2018; 11:7493-7501. [PMID: 30498362 PMCID: PMC6207221 DOI: 10.2147/ott.s161852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Family with sequence similarity 129, member B (FAM129B), also called MINERVA, is upregulated and promotes tumor invasion in multiple types of cancer. However, the mechanism and clinicopathological significance of FAM129B remains unclear. Materials and methods Online KM-plotter tool and immunohistochemistry were used to predict the prognostic value of FAM129B expression in lung cancer tissues. Western blotting analysis, MTT, colony formation assay and matrigel invasion assay were performed after overexpressing or depleting FAM129B. Results In this study, using the online KM-plotter tool, we found FAM129B was correlated with adverse outcome in non-small cell lung cancer (NSCLC) patients (P<0.001). Immunohistochemistry results revealed that FAM129B showed negative or dim expression in normal lung tissues while presented positive cytoplasmic expression in both squamous cell lung carcinoma and lung adenocarcinoma. The positive ratio of FAM129B in clinical NSCLC tissue samples (77/187, 41.2%) was significantly higher than that in normal lung tissue samples (8/68, 11.8%; P<0.001). FAM129B expression associated with advanced TNM staging (P<0.001) and positive regional lymph node metastasis (P<0.001). The results of Kaplan-Meier analysis suggested that the survival time of patients with positive FAM129B expression was significantly shorter than those with negatively FAM129B expression (P<0.001). Proliferation and invasion assay revealed that FAM129B prominently facilitated tumor proliferation and invasion in NSCLC cells. Western blotting results revealed that FAM129B upregulated the expression of MMP2 and Cyclin D1 by enhancing the phosphorylation of FAK at Tyr 397 and Tyr 925. Incorporation of FAK inhibitor in the medium significantly downregulated the phosphorylation of FAK and subsequently attenuated increasing expression of MMP2 and Cyclin D1 induced by FAM129B overexpression. Conclusion Our results indicated that FAM129B may be a new prognosis predictor of NSCLC patients and impact tumor invasion and proliferation of NSCLC cells through promoting the activation of FAK signaling.
Collapse
Affiliation(s)
- Xiaoming Zhou
- Department of Respiratory Medicine, The Shengjing Hospital of China Medical University, Shenyang, China
| | - Fangfei Yang
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Qin Zhang
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Miao
- Department of Pathology, The First Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xuejun Hu
- Department of Respiratory Disease in Geratology, The First Hospital of China Medical University, Shenyang, China
| | - Ailin Li
- Department of Radiotherapy, The First Hospital of China Medical University, Shenyang, China,
| | - Gang Hou
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Qiuyue Wang
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Respiratory Medicine, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
A phase 2 study of vorinostat in locally advanced, recurrent, or metastatic adenoid cystic carcinoma. Oncotarget 2018; 8:32918-32929. [PMID: 28415633 PMCID: PMC5464838 DOI: 10.18632/oncotarget.16464] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023] Open
Abstract
Purpose Vorinostat is a histone deacetylase inhibitor (HDACi). Based on a confirmed partial response (PR) in an adenoid cystic carcinoma (ACC) patient treated with vorinostat in a prior phase 1 trial, we initiated this phase 2 trial. Methods: Vorinostat was administered orally 400 mg daily, 28 day cycles. The primary objective was to evaluate response rate (RR). Exploratory studies included whole exome sequencing (WES) of selected patients. Results Thirty patients were enrolled. Median age of patients was 53 years (range 21–73). Median number of cycles was 5 (range 1-66). Lymphopenia (n = 5), hypertension (n = 3), oral pain (n = 2), thromboembolic events (n = 2) and fatigue (n = 2) were the only grade 3 adverse events (AEs) that occurred in more than 1 patient. Eleven patients were dose reduced secondary to drug-related AEs. Two patients had a partial response (PR), with response durations of 53 and 7.2 months. One patient had a minor response with a decrease in ascites (for 19 cycles). Stable disease was the best response in 27 patients. Targeted and WES of 8 patients in this trial identified mutations in chromatin remodeling genes highlighting the role of the epigenome in ACC. Conclusion: Vorinostat demonstrated efficacy in patients with ACC supporting the inclusion of HDACi in future studies to treat ACC.
Collapse
|
21
|
Gusnanto A, Gosling JP, Pope C. Identification of transcript regulatory patterns in cell differentiation. Bioinformatics 2017; 33:3235-3242. [PMID: 28655167 DOI: 10.1093/bioinformatics/btx406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 06/21/2017] [Indexed: 11/13/2022] Open
Abstract
Motivation Studying transcript regulatory patterns in cell differentiation is critical in understanding its complex nature of the formation and function of different cell types. This is done usually by measuring gene expression at different stages of the cell differentiation. However, if the gene expression data available are only from the mature cells, we have some challenges in identifying transcript regulatory patterns that govern the cell differentiation. Results We propose to exploit the information of the lineage of cell differentiation in terms of correlation structure between cell types. We assume that two different cell types that are close in the lineage will exhibit many common genes that are co-expressed relative to those that are far in the lineage. Current analysis methods tend to ignore this correlation by testing for differential expression assuming some sort of independence between cell types. We employ a Bayesian approach to estimate the posterior distribution of the mean of expression in each cell type, by taking into account the cell formation path in the lineage. This enables us to infer genes that are specific in each cell type, indicating the genes are involved in directing the cell differentiation to that particular cell type. We illustrate the method using gene expression data from a study of haematopoiesis. Availability and implementation R codes to perform the analysis are available in http://www1.maths.leeds.ac.uk/∼arief/R/CellDiff/. Contact a.gusnanto@leeds.ac.uk. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Arief Gusnanto
- Department of Statistics, University of Leeds, Leeds LS2 9JT, UK
| | | | - Christopher Pope
- Department of Statistics, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
22
|
Doll S, Urisman A, Oses-Prieto JA, Arnott D, Burlingame AL. Quantitative Proteomics Reveals Fundamental Regulatory Differences in Oncogenic HRAS and Isocitrate Dehydrogenase (IDH1) Driven Astrocytoma. Mol Cell Proteomics 2017; 16:39-56. [PMID: 27834733 PMCID: PMC5217781 DOI: 10.1074/mcp.m116.063883] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/04/2016] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiformes (GBMs) are high-grade astrocytomas and the most common brain malignancies. Primary GBMs are often associated with disturbed RAS signaling, and expression of oncogenic HRAS results in a malignant phenotype in glioma cell lines. Secondary GBMs arise from lower-grade astrocytomas, have slower progression than primary tumors, and contain IDH1 mutations in over 70% of cases. Despite significant amount of accumulating genomic and transcriptomic data, the fundamental mechanistic differences of gliomagenesis in these two types of high-grade astrocytoma remain poorly understood. Only a few studies have attempted to investigate the proteome, phosphorylation signaling, and epigenetic regulation in astrocytoma. In the present study, we applied quantitative phosphoproteomics to identify the main signaling differences between oncogenic HRAS and mutant IDH1-driven glioma cells as models of primary and secondary GBM, respectively. Our analysis confirms the driving roles of the MAPK and PI3K/mTOR signaling pathways in HRAS driven cells and additionally uncovers dysregulation of other signaling pathways. Although a subset of the signaling changes mediated by HRAS could be reversed by a MEK inhibitor, dual inhibition of MEK and PI3K resulted in more complete reversal of the phosphorylation patterns produced by HRAS expression. In contrast, cells expressing mutant IDH1 did not show significant activation of MAPK or PI3K/mTOR pathways. Instead, global downregulation of protein expression was observed. Targeted proteomic analysis of histone modifications identified significant histone methylation, acetylation, and butyrylation changes in the mutant IDH1 expressing cells, consistent with a global transcriptional repressive state. Our findings offer novel mechanistic insight linking mutant IDH1 associated inhibition of histone demethylases with specific histone modification changes to produce global transcriptional repression in secondary glioblastoma. Our proteomic datasets are available for download and provide a comprehensive catalogue of alterations in protein abundance, phosphorylation, and histone modifications in oncogenic HRAS and IDH1 driven astrocytoma cells beyond the transcriptomic level.
Collapse
Affiliation(s)
- Sophia Doll
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - Anatoly Urisman
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - Juan A Oses-Prieto
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - David Arnott
- §Department of Protein Chemistry, Genentech Inc, South San Francisco, 94158-2517 California
| | - Alma L Burlingame
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California;
| |
Collapse
|
23
|
Patel SJ, Trivedi GL, Darie CC, Clarkson BD. The possible roles of B-cell novel protein-1 (BCNP1) in cellular signalling pathways and in cancer. J Cell Mol Med 2016; 21:456-466. [PMID: 27680505 PMCID: PMC5323820 DOI: 10.1111/jcmm.12989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 08/12/2016] [Indexed: 01/15/2023] Open
Abstract
B‐cell novel protein‐1 (BCNP1) or Family member of 129C (FAM129C) was identified as a B‐cell‐specific plasma‐membrane protein. Bioinformatics analysis predicted that BCNP1 might be heavily phosphorylated. The BCNP1 protein contains a pleckstrin homology (PH) domain, two proline‐rich (PR) regions and a Leucine Zipper (LZ) domain suggesting that it may be involved in protein‐protein interactions. Using The Cancer Genome Atlas (TCGA) data sets, we investigated the correlation of alteration of the BCNP1 copy‐number changes and mutations in several cancer types. We also investigated the function of BCNP1 in cellular signalling pathways. We found that BCNP1 is highly altered in some types of cancers and that BCNP1 copy‐number changes and mutations co‐occur with other molecular alteration events for TP53 (tumour protein P53), PIK3CA (Phosphatidylinositol‐4,5‐Bisphosphate 3‐Kinase, Catalytic Subunit Alpha), MAPK1 (mitogen‐activated protein kinase‐1; ERK: extracellular signal regulated kinase), KRAS (Kirsten rat sarcoma viral oncogene homolog) and AKT2 (V‐Akt Murine Thymoma Viral Oncogene Homolog 2). We also found that PI3K (Phoshoinositide 3‐kinase) inhibition and p38 MAPK (p38 mitogen‐activated protein kinase) activation leads to reduction in phosphorylation of BCNP1 at serine residues, suggesting that BCNP1 phosphorylation is PI3K and p38MAPK dependent and that it might be involved in cancer. Its degradation depends on a proteasome‐mediated pathway.
Collapse
Affiliation(s)
- Sapan J Patel
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology and Chemistry Program, New York, NY, USA.,Department of Chemistry and Biomolecular Science, Clarkson University, Biochemistry and Proteomics Group, Potsdam, NY, USA
| | | | - Costel C Darie
- Department of Chemistry and Biomolecular Science, Clarkson University, Biochemistry and Proteomics Group, Potsdam, NY, USA
| | - Bayard D Clarkson
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology and Chemistry Program, New York, NY, USA
| |
Collapse
|
24
|
Abstract
Ras GTPase-activating proteins (GAPs) are important regulators for Ras activation, which is instrumental in tumor development. However, the mechanism underlying this regulation remains elusive. We demonstrate here that activated EGFR phosphorylates the Y593 residue of the protein known as family with sequence similarity 129, member B (FAM129B), which is overexpressed in many types of human cancer. FAM129B phosphorylation increased the interaction between FAM129B and Ras, resulting in reduced binding of p120-RasGAP to Ras. FAM129B phosphorylation promoted Ras activation, increasing ERK1/2- and PKM2-dependent β-catenin transactivation and leading to the enhanced glycolytic gene expression and the Warburg effect; promoting tumor cell proliferation and invasion; and supporting brain tumorigenesis. Our studies unearthed a novel and important mechanism underlying EGFR-mediated Ras activation in tumor development.
Collapse
|
25
|
Tanaka T, Iino M, Goto K. Knockdown of Sec8 enhances the binding affinity of c-Jun N-terminal kinase (JNK)-interacting protein 4 for mitogen-activated protein kinase kinase 4 (MKK4) and suppresses the phosphorylation of MKK4, p38, and JNK, thereby inhibiting apoptosis. FEBS J 2014; 281:5237-50. [PMID: 25244576 DOI: 10.1111/febs.13063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/15/2014] [Accepted: 09/18/2014] [Indexed: 01/19/2023]
Abstract
The exocyst complex, also called the Sec6/8 complex, is important for targeting exocytic vesicles to specific docking sites on the plasma membrane in yeast and mammalian cells. In addition to these original findings, recent results of studies suggest that Sec8 is also involved in oncogenesis, although the functional implications of Sec8 in cancer cells are not well understood. c-Jun N-terminal kinase-interacting protein 4 (JIP4) is a scaffold protein that plays a crucial role in the regulation of mitogen-activated protein kinase (MAPK) signaling cascades. The present study examined how Sec8 is involved in JIP4-mediated MAPK signaling under apoptotic conditions. It was found that Sec8 binds to and regulates JIP4, and that knockdown of Sec8 enhances the binding of JIP4 to MAPK kinase 4, thereby decreasing the phosphorylation of MAPK kinase 4, JNK, and p38. These results raise the possibility that Sec8 serves as an important regulator of MAPK signaling cascades.
Collapse
Affiliation(s)
- Toshiaki Tanaka
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Japan; Department of Dentistry, Oral and Maxillofacial Surgery, Plastic and Reconstructive Surgery, Yamagata University School of Medicine, Japan
| | | | | |
Collapse
|
26
|
Conrad W, Major MB, Cleary MA, Ferrer M, Roberts B, Marine S, Chung N, Arthur WT, Moon RT, Berndt JD, Chien AJ. FAM129B is a novel regulator of Wnt/β-catenin signal transduction in melanoma cells. F1000Res 2013; 2:134. [PMID: 24358901 PMCID: PMC3829391 DOI: 10.12688/f1000research.2-134.v2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2013] [Indexed: 01/08/2023] Open
Abstract
The inability of targeted BRAF inhibitors to produce long-lasting improvement in the clinical outcome of melanoma highlights a need to identify additional approaches to inhibit melanoma growth. Recent studies have shown that activation of the Wnt/β-catenin pathway decreases tumor growth and cooperates with ERK/MAPK pathway inhibitors to promote apoptosis in melanoma. Therefore, the identification of Wnt/β-catenin regulators may advance the development of new approaches to treat this disease. In order to move towards this goal we performed a large scale small-interfering RNA (siRNA) screen for regulators of β-catenin activated reporter activity in human HT1080 fibrosarcoma cells. Integrating large scale siRNA screen data with phosphoproteomic data and bioinformatics enrichment identified a protein, FAM129B, as a potential regulator of Wnt/β-catenin signaling. Functionally, we demonstrated that siRNA-mediated knockdown of FAM129B in A375 and A2058 melanoma cell lines inhibits WNT3A-mediated activation of a β-catenin-responsive luciferase reporter and inhibits expression of the endogenous Wnt/β-catenin target gene, AXIN2. We also demonstrate that FAM129B knockdown inhibits apoptosis in melanoma cells treated with WNT3A. These experiments support a role for FAM129B in linking Wnt/β-catenin signaling to apoptosis in melanoma.
Collapse
Affiliation(s)
- Willliam Conrad
- The Howard Hughes Medical Institute, Seattle WA, 98195, USA ; Department of Pharmacology, University of Washington School of Medicine, Seattle WA, 98195, USA
| | - Michael B Major
- The Howard Hughes Medical Institute, Seattle WA, 98195, USA ; Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7295, USA
| | - Michele A Cleary
- Rosetta Inpharmatics, LLC, Merck & Co Inc., Seattle WA, 98109, USA ; Merck & Co., Inc., West Point PA, 19486, USA
| | - Marc Ferrer
- Department of Automated Biotechnology, Merck, North Wales PA, 19454, USA ; NCATS/NIH, Rockville MD, 20850, USA
| | - Brian Roberts
- Rosetta Inpharmatics, LLC, Merck & Co Inc., Seattle WA, 98109, USA ; Hudson Alpha Institute for Biotechnology, Huntsville AL, 35806, USA
| | - Shane Marine
- Department of Screening and Protein Sciences, Merck Research Laboratories, North Wales PA, 19454, USA
| | - Namjin Chung
- Rosetta Inpharmatics, LLC, Merck & Co Inc., Seattle WA, 98109, USA ; Bristol-Myers Squibb Company, Applied Genomics, Princeton NJ, 08543, USA
| | - William T Arthur
- Rosetta Inpharmatics, LLC, Merck & Co Inc., Seattle WA, 98109, USA ; Seattle Genetics, Bothell WA, 98021, USA
| | - Randall T Moon
- The Howard Hughes Medical Institute, Seattle WA, 98195, USA ; Department of Pharmacology, University of Washington School of Medicine, Seattle WA, 98195, USA ; The Institute for Stem Cell and Regenerative Medicine, Seattle WA, 98109, USA
| | - Jason D Berndt
- The Howard Hughes Medical Institute, Seattle WA, 98195, USA ; Department of Pharmacology, University of Washington School of Medicine, Seattle WA, 98195, USA ; The Institute for Stem Cell and Regenerative Medicine, Seattle WA, 98109, USA
| | - Andy J Chien
- Division of Dermatology, Department of Medicine, University of Washington, Seattle WA, 98195, USA ; The Institute for Stem Cell and Regenerative Medicine, Seattle WA, 98109, USA
| |
Collapse
|
27
|
Miller NH, Justice CM, Marosy B, Swindle K, Kim Y, Roy-Gagnon MH, Sung H, Behneman D, Doheny KF, Pugh E, Wilson AF. Intra-familial tests of association between familial idiopathic scoliosis and linked regions on 9q31.3-q34.3 and 16p12.3-q22.2. Hum Hered 2012; 74:36-44. [PMID: 23154503 PMCID: PMC4123546 DOI: 10.1159/000343751] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 09/07/2012] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Custom genotyping of markers in families with familial idiopathic scoliosis were used to fine-map candidate regions on chromosomes 9 and 16 in order to identify candidate genes that contribute to this disorder and prioritize them for next-generation sequence analysis. METHODS Candidate regions on 9q and 16p-16q, previously identified as linked to familial idiopathic scoliosis in a study of 202 families, were genotyped with a high-density map of single nucleotide polymorphisms. Tests of linkage for fine-mapping and intra-familial tests of association, including tiled regression, were performed on scoliosis as both a qualitative and quantitative trait. RESULTS AND CONCLUSIONS Nominally significant linkage results were found for markers in both candidate regions. Results from intra-familial tests of association and tiled regression corroborated the linkage findings and identified possible candidate genes suitable for follow-up with next-generation sequencing in these same families. Candidate genes that met our prioritization criteria included FAM129B and CERCAM on chromosome 9 and SYT1, GNAO1, and CDH3 on chromosome 16.
Collapse
Affiliation(s)
- Nancy H Miller
- Department of Orthopaedic Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Oishi H, Itoh S, Matsumoto K, Ishitobi H, Suzuki R, Ema M, Kojima T, Uchida K, Kato M, Miyata T, Takahashi S. Delayed cutaneous wound healing in Fam129b/Minerva-deficient mice. J Biochem 2012; 152:549-55. [PMID: 22977259 DOI: 10.1093/jb/mvs100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
A recent integrative analysis using a phosphoproteomic approach identified FAM129B, also known as MINERVA, as a downstream effector of the MAP kinase pathway in human melanoma cells. FAM129B protein, which is a member of a small family of proteins, was also found to suppress TNFα/cycloheximide-induced apoptosis in HeLa cells. To investigate the physiological functions of Fam129b in vivo, we generated gene-targeted Fam129b-mutant mice in which, the amino terminal coding exon was replaced by lacZ. We found that homozygous mutant mice are viable and fertile and that Fam129b is considerably expressed in most of the epidermal keratinocytes of both embryonic and adult mice. Although no gross defect was observed in the skin of the Fam129b-deficient mice, wound healing subsequent to skin puncturing was significantly delayed. Furthermore, overexpression of Fam129b promoted HaCaT cell motility in an N-terminal pleckstrin homology domain-dependent manner, but not proliferation. Microarray analysis of the Fam129b transfectant exhibited substantial upregulation of several genes related to wound repair and cell motility. These results suggest that expression of Fam129b in epidermal keratinocytes accompanied by alteration of wound healing-related gene expression is necessary for regulation of cell motility and thereby, contributes to the appropriate wound healing process.
Collapse
Affiliation(s)
- Hisashi Oishi
- Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba 305-8575, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lacruz RS, Smith CE, Bringas P, Chen YB, Smith SM, Snead ML, Kurtz I, Hacia JG, Hubbard MJ, Paine ML. Identification of novel candidate genes involved in mineralization of dental enamel by genome-wide transcript profiling. J Cell Physiol 2012; 227:2264-75. [PMID: 21809343 DOI: 10.1002/jcp.22965] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The gene repertoire regulating vertebrate biomineralization is poorly understood. Dental enamel, the most highly mineralized tissue in mammals, differs from other calcifying systems in that the formative cells (ameloblasts) lack remodeling activity and largely degrade and resorb the initial extracellular matrix. Enamel mineralization requires that ameloblasts undergo a profound functional switch from matrix-secreting to maturational (calcium transport, protein resorption) roles as mineralization progresses. During the maturation stage, extracellular pH decreases markedly, placing high demands on ameloblasts to regulate acidic environments present around the growing hydroxyapatite crystals. To identify the genetic events driving enamel mineralization, we conducted genome-wide transcript profiling of the developing enamel organ from rat incisors and highlight over 300 genes differentially expressed during maturation. Using multiple bioinformatics analyses, we identified groups of maturation-associated genes whose functions are linked to key mineralization processes including pH regulation, calcium handling, and matrix turnover. Subsequent qPCR and Western blot analyses revealed that a number of solute carrier (SLC) gene family members were up-regulated during maturation, including the novel protein Slc24a4 involved in calcium handling as well as other proteins of similar function (Stim1). By providing the first global overview of the cellular machinery required for enamel maturation, this study provide a strong foundation for improving basic understanding of biomineralization and its practical applications in healthcare.
Collapse
Affiliation(s)
- Rodrigo S Lacruz
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|