1
|
Milia S, Leonard AS, Mapel XM, Bernal Ulloa SM, Drögemüller C, Pausch H. Taurine pangenome uncovers a segmental duplication upstream of KIT associated with depigmentation in white-headed cattle. Genome Res 2025; 35:1041-1052. [PMID: 39694857 PMCID: PMC12047182 DOI: 10.1101/gr.279064.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Cattle have been selectively bred for coat color, spotting, and depigmentation patterns. The assumed autosomal dominant inherited genetic variants underlying the characteristic white head of Fleckvieh, Simmental, and Hereford cattle have not been identified yet, although the contribution of structural variation upstream of the KIT gene has been proposed. Here, we construct a graph pangenome from 24 haplotype assemblies representing seven taurine cattle breeds to identify and characterize the white-head-associated locus for the first time based on long-read sequencing data and pangenome analyses. We introduce a pangenome-wide association mapping approach that examines assembly path similarities within the graph to reveal an association between two most likely serial alleles of a complex structural variant (SV) 66 kb upstream of KIT and facial depigmentation. The complex SV contains a variable number of tandemly duplicated 14.3 kb repeats, consisting of LTRs, LINEs, and other repetitive elements, leading to misleading alignments of short and long reads when using a linear reference. We align 250 short-read sequencing samples spanning 15 cattle breeds to the pangenome graph, further validating that the alleles of the SV segregate with head depigmentation. We estimate an increased count of repeats in Hereford relative to Simmental and other white-headed cattle breeds from the graph alignment coverage, suggesting a large under-assembly in the current Hereford-based cattle reference genome, which had fewer copies. Our work shows that exploiting assembly path similarities within graph pangenomes can reveal trait-associated complex SVs.
Collapse
Affiliation(s)
- Sotiria Milia
- Animal Genomics, ETH Zurich, Zurich 8092, Switzerland
| | | | | | | | - Cord Drögemüller
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern 3012, Switzerland
| | - Hubert Pausch
- Animal Genomics, ETH Zurich, Zurich 8092, Switzerland;
| |
Collapse
|
2
|
Kabirova E, Ryzhkova A, Lukyanchikova V, Khabarova A, Korablev A, Shnaider T, Nuriddinov M, Belokopytova P, Smirnov A, Khotskin NV, Kontsevaya G, Serova I, Battulin N. TAD border deletion at the Kit locus causes tissue-specific ectopic activation of a neighboring gene. Nat Commun 2024; 15:4521. [PMID: 38806452 PMCID: PMC11133455 DOI: 10.1038/s41467-024-48523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
Topologically associated domains (TADs) restrict promoter-enhancer interactions, thereby maintaining the spatiotemporal pattern of gene activity. However, rearrangements of the TADs boundaries do not always lead to significant changes in the activity pattern. Here, we investigated the consequences of the TAD boundaries deletion on the expression of developmentally important genes encoding tyrosine kinase receptors: Kit, Kdr, Pdgfra. We used genome editing in mice to delete the TADs boundaries at the Kit locus and characterized chromatin folding and gene expression in pure cultures of fibroblasts, mast cells, and melanocytes. We found that although Kit is highly active in both mast cells and melanocytes, deletion of the TAD boundary between the Kit and Kdr genes results in ectopic activation only in melanocytes. Thus, the epigenetic landscape, namely the mutual arrangement of enhancers and actively transcribing genes, is important for predicting the consequences of the TAD boundaries removal. We also found that mice without a TAD border between the Kit and Kdr genes have a phenotypic manifestation of the mutation - a lighter coloration. Thus, the data obtained shed light on the principles of interaction between the 3D chromatin organization and epigenetic marks in the regulation of gene activity.
Collapse
Affiliation(s)
- Evelyn Kabirova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | | | | | - Anna Khabarova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Alexey Korablev
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | | | | | - Polina Belokopytova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | | | | | | | - Irina Serova
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Nariman Battulin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia.
- Novosibirsk State University, Novosibirsk, Russia.
| |
Collapse
|
3
|
Fuda S, Angelini DF, Colopi A, Guida E, Onorato A, Grimaldi P, Travaglini S, Jannini EA, Dolci S. A minimal promoter region of Kit gene recapitulates mast cell differentiation in development, aging and inflammation. Mech Ageing Dev 2023; 212:111820. [PMID: 37178832 DOI: 10.1016/j.mad.2023.111820] [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: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023]
Abstract
To follow mast cells (MCs) distribution during aging and inflammation, we characterized two transgenic mouse models in which the EGFP expression is controlled by 9kb or 12kb of Kit gene promoter, defined as p18 and p70, respectively. We detected EGFP-positive cells in the serosal surfaces of the peritoneum, pleuras and pericardium, mucosal cavities, and connective tissue of almost all organs including gonads of p70, but not of p18 mice. By FACS and immunofluorescence for FcεR1, Kit and β7-integrin, we found that these EGFP positive cells were MCs. In non-inflammatory conditions, a higher percentage of EGFP positive cells was found in juvenile with respect to adult serosal surfaces, but no differences between males and females at both developmental ages. We found, however, a striking difference in developing gonads, with low numbers of EGFP positive cells in foetal ovaries compared to age matched testes. Under inflammatory conditions caused by high fat diet (HFD), mice showed an increase in serosal EGFP positve cells. Altogether our results identify a regulatory region of the Kit gene, activated in MCs and that directing EGFP expression, can be employed to trace this immune cell type throughout the organism and in different animal conditions.
Collapse
Affiliation(s)
- Serena Fuda
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Daniela F Angelini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Ambra Colopi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata
| | - Eugenia Guida
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Angelo Onorato
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Paola Grimaldi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Serena Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Emmanuele A Jannini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| |
Collapse
|
4
|
Luo X, Chen J, Yang H, Hu X, Alphonse MP, Shen Y, Kawakami Y, Zhou X, Tu W, Kawakami T, Wan M, Archer NK, Wang H, Gao P. Dendritic cell immunoreceptor drives atopic dermatitis by modulating oxidized CaMKII-involved mast cell activation. JCI Insight 2022; 7:152559. [PMID: 35113811 PMCID: PMC8983143 DOI: 10.1172/jci.insight.152559] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Allergens have been identified as potential triggers in patients with atopic dermatitis (AD). AD patients are highly sensitive to cockroach allergen. The underlying mechanism, however, remains undetermined. Here, we established a cockroach allergen-induced AD-like mouse model and demonstrated that repeated exposure to cockroach allergen led to aggravated mouse skin inflammation, characterized by increased type 2 immunity, type 2 innate lymphoid cells (ILC2s), and mast cells. Increased skin mast cells were also observed in AD patients. AD mice with mast cell-deficient mice (kitW-sh/W-sh) showed diminished skin inflammation, suggesting that mast cells are required in allergen-induced skin inflammation. Furthermore, dendritic cell immuno-receptor (DCIR) is up-regulated in skin mast cells of AD patients and mediates allergen binding and uptake. DCIR-/- mice or reconstituted kitW-sh/W-sh mice with DCIR-/- mast cells showed a significant reduction in AD-like inflammation. Both in vitro and in vivo analyses demonstrated that DCIR-/- mast cells had reduced IgE-mediated mast cell activation and passive cutaneous anaphylaxis. Mechanistically, DCIR regulates allergen-induced IgE-mediated mast cell ROS generation and oxidation of calmodulin kinase II (ox-CaMKII). ROS-resistant CaMKII (MM-VVδ) prevents allergen-induced mast cell activation and inflammatory mediator release. Our study reveals a previously unrecognized DCIR-ROS-CaMKII axis that controls allergen-induced mast cell activation and AD-like inflammation.
Collapse
Affiliation(s)
- Xiaoyan Luo
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Jingsi Chen
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Huan Yang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yuko Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Xiaoying Zhou
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Mei Wan
- Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Hua Wang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| |
Collapse
|
5
|
Jain R, Tikoo S, On K, Martinez B, Dervish S, Cavanagh LL, Weninger W. Visualizing murine breast and melanoma tumor microenvironment using intravital multiphoton microscopy. STAR Protoc 2021; 2:100722. [PMID: 34458865 PMCID: PMC8379651 DOI: 10.1016/j.xpro.2021.100722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Intravital multiphoton imaging of the tumor milieu allows for the dissection of intricate and dynamic biological processes in situ. Herein, we present a step-by-step protocol for setting up an experimental cancer imaging model that has been optimized for solid tumors such as breast cancer and melanoma implanted in the flanks of mice. This protocol can be utilized for dissecting tumor-immune cell dynamics in vivo or other tumor-specific biological questions. For complete details on the use of this protocol for intravital imaging of breast cancer, please refer to Tikoo et al. (2021a), and for intravital imaging of melanoma, please refer to Tikoo et al. (2021b). Detailed protocol for setting up high-resolution intravital imaging of murine tumors 3D printing of custom stage inserts for tumor stabilization Procedures for cannulation of blood vessels Surgical preparation and tissue stabilization for imaging tumor milieu in vivo
Collapse
Affiliation(s)
- Rohit Jain
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Shweta Tikoo
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Kathy On
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Brendon Martinez
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Suat Dervish
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Lois L Cavanagh
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Wolfgang Weninger
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia.,Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| |
Collapse
|
6
|
Jachetti E, D'Incà F, Danelli L, Magris R, Dal Secco C, Vit F, Cancila V, Tripodo C, Scapini P, Colombo MP, Pucillo C, Frossi B. Frontline Science: Mast cells regulate neutrophil homeostasis by influencing macrophage clearance activity. J Leukoc Biol 2019; 105:633-644. [PMID: 30817046 DOI: 10.1002/jlb.4hi1018-390r] [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: 10/13/2018] [Revised: 01/13/2019] [Accepted: 01/29/2019] [Indexed: 12/25/2022] Open
Abstract
The receptor tyrosine kinase cKit and its ligand stem cell factor are essential for mast cells (MC) development and survival. Strains with mutations affecting the Kit gene display a profound MC deficiency in all tissues and have been extensively used to investigate the role of MC in both physiologic and pathologic conditions. However, these mice present a variety of abnormalities in other immune cell populations that can affect the interpretation of MC-related responses. C57BL/6 KitW-sh are characterized by an aberrant extramedullary myelopoiesis and systemic neutrophilia. MC deficiency in KitW-sh mice can be selectively repaired by engraftment with in vitro-differentiated MC to validate MC-specific functions. Nevertheless, the impact of MC reconstitution on other immune populations has never been evaluated in detail. Here, we specifically investigated the neutrophil compartment in primary and secondary lymphoid organs of C57BL/6 KitW-sh mice before and after MC reconstitution. We found that, albeit not apparently affecting neutrophils phenotype or maturation, MC reconstitution of KitW-sh mice restored the number of neutrophils at a level similar to that of wild-type C57BL/6 mice. In vitro and ex vivo experiments indicated that MC can influence neutrophil clearance by increasing macrophages' phagocytic activity. Furthermore, the G-CSF/IL-17 axis was also influenced by the presence or absence of MC in KitW-sh mice. These data suggest that MC play a role in the control of neutrophil homeostasis and that this aspect should be taken into account in the interpretation of results obtained using KitW-sh mice.
Collapse
Affiliation(s)
- Elena Jachetti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica D'Incà
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Luca Danelli
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Raffaella Magris
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Chiara Dal Secco
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Filippo Vit
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Patrizia Scapini
- Division of General Pathology, University of Verona, Verona, Italy
| | - Mario Paolo Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Pucillo
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| | - Barbara Frossi
- Immunology Section, Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
7
|
Todaro F, Campolo F, Barrios F, Pellegrini M, Di Cesare S, Tessarollo L, Rossi P, Jannini EA, Dolci S. Regulation of Kit Expression in Early Mouse Embryos and ES Cells. Stem Cells 2019; 37:332-344. [PMID: 30566254 DOI: 10.1002/stem.2960] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 11/15/2018] [Accepted: 11/24/2018] [Indexed: 01/01/2023]
Abstract
Kit is a growth factor receptor that regulates proliferation and/or survival of many embryonic and postnatal stem cell types. When mutated, it can induce malignant transformation of the host cells. To dissect the Kit role in the control of ESC pluripotency, we studied its expression during early mouse embryogenesis and during the process of ESC derivation from inner cell mass (ICM) cells. We followed the in vitro development of early mouse embryos obtained from transgenic mice carrying Kit promoter regions fused to EGFP (Kit-EGFP) and found that they initiate EGFP expression at morula stage. EGFP expression is then maintained in the blastocyst, within the ICM, and its levels increase when cultured in the presence of MAPK and GSK3β inhibitors (2i) plus LIF compared with the LIF-only condition. Kit-EGFP ESCs showed nonhomogeneous EGFP expression pattern when cultured in LIF condition, but they upregulated EGFP expression, as well as that of Sox2, Nanog, Prdm14, when shifted to 2i-LIF culture. Similarly, primordial germ cells (PGCs) in the process of embryonic germ cell (EGC) conversion showed enhanced EGFP expression in 2i-LIF. Kit expression was affected by manipulating Sox2 levels in ESCs. Chromatin immunoprecipitation experiments confirmed that Sox2 binds Kit regulatory regions containing Sox2 consensus sequences. Finally, Kit constitutive activation induced by the D814Y mutation increased ESC proliferation and cloning efficiency in vitro and in teratoma assays in vivo. Our results identify Kit as a pluripotency-responsive gene and suggest a role for Kit in the regulation of ESC proliferation. Stem Cells 2019;37:332-344.
Collapse
Affiliation(s)
- Federica Todaro
- Dipartimento di Biomedicina e Prevenzione, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Federica Campolo
- Dipartimento di Biomedicina e Prevenzione, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Florencia Barrios
- Dipartimento di Biomedicina e Prevenzione, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | | | - Silvia Di Cesare
- Dipartimento di Medicina dei Sistemi, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Lino Tessarollo
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Pellegrino Rossi
- Dipartimento di Biomedicina e Prevenzione, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Emmanuele A Jannini
- Dipartimento di Medicina dei Sistemi, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Susanna Dolci
- Dipartimento di Biomedicina e Prevenzione, Università degli Studi di Roma Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Tikoo S, Barki N, Jain R, Zulkhernain NS, Buhner S, Schemann M, Weninger W. Imaging of mast cells. Immunol Rev 2019; 282:58-72. [PMID: 29431206 DOI: 10.1111/imr.12631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells are a part of the innate immune system implicated in allergic reactions and the regulation of host-pathogen interactions. The distribution, morphology and biochemical composition of mast cells has been studied in detail in vitro and on tissue sections both at the light microscopic and ultrastructural level. More recently, the development of fluorescent reporter strains and intravital imaging modalities has enabled first glimpses of the real-time behavior of mast cells in situ. In this review, we describe commonly used imaging approaches to study mast cells in cell culture as well as within normal and diseased tissues. We further describe the interrogation of mast cell function via imaging by providing a detailed description of mast cell-nerve plexus interactions in the intestinal tract. Together, visualizing mast cells has expanded our view of these cells in health and disease.
Collapse
Affiliation(s)
- Shweta Tikoo
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia
| | - Natasja Barki
- LS Human Biology, Technical University München, München, Germany
| | - Rohit Jain
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia
| | | | - Sabine Buhner
- LS Human Biology, Technical University München, München, Germany
| | - Michael Schemann
- LS Human Biology, Technical University München, München, Germany
| | - Wolfgang Weninger
- The Centenary Institute, Newtown, NSW, Australia.,Discipline of Dermatology, Sydney Medical School, Sydney, NSW, Australia.,Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
9
|
Berrozpe G, Bryant GO, Warpinski K, Spagna D, Narayan S, Shah S, Ptashne M. Polycomb Responds to Low Levels of Transcription. Cell Rep 2018; 20:785-793. [PMID: 28746865 DOI: 10.1016/j.celrep.2017.06.076] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/14/2017] [Accepted: 06/23/2017] [Indexed: 01/27/2023] Open
Abstract
How is Polycomb (Pc), a eukaryotic negative regulator of transcription, targeted to specific mammalian genes? Our genome-wide analysis of the Pc mark H3K27me3 in murine cells revealed that Pc is preferentially associated with CpG island promoters of genes that are transcribed at a low level and less so with promoters of genes that are either silent or more highly expressed. Studies of the CpG island promoter of the Kit gene demonstrate that Pc is largely absent when the gene is silent in myeloid cells, as well as when the gene is highly expressed in mast cells. Manipulations that increase transcription in the former case, and reduce it in the latter, increase Pc occupancy. The average negative effect of Pc, we infer, is about 2-fold. We suggest possible biological roles for such negative effects and propose a mechanism by which Pc might be recruited to weakly transcribed genes.
Collapse
Affiliation(s)
- Georgina Berrozpe
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Gene O Bryant
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Katherine Warpinski
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Dan Spagna
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Santosh Narayan
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Shivangi Shah
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Mark Ptashne
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA.
| |
Collapse
|
10
|
Narayan S, Bryant G, Shah S, Berrozpe G, Ptashne M. OCT4 and SOX2 Work as Transcriptional Activators in Reprogramming Human Fibroblasts. Cell Rep 2018; 20:1585-1596. [PMID: 28813671 DOI: 10.1016/j.celrep.2017.07.071] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 01/06/2023] Open
Abstract
SOX2 and OCT4, in conjunction with KLF4 and cMYC, are sufficient to reprogram human fibroblasts to induced pluripotent stem cells (iPSCs), but it is unclear if they function as transcriptional activators or as repressors. We now show that, like OCT4, SOX2 functions as a transcriptional activator. We substituted SOX2-VP16 (a strong activator) for wild-type (WT) SOX2, and we saw an increase in the efficiency and rate of reprogramming, whereas the SOX2-HP1 fusion (a strong repressor) eliminated reprogramming. We report that, at an early stage of reprogramming, virtually all DNA-bound OCT4, SOX2, and SOX2-VP16 were embedded in putative enhancers, about half of which were created de novo. Those associated with SOX2-VP16 were, on average, stronger than those bearing WT SOX2. Many newly created putative enhancers were transient, and many transcription factor locations on DNA changed as reprogramming progressed. These results are consistent with the idea that, during reprogramming, there is an intermediate state that is distinct from both parental cells and iPSCs.
Collapse
Affiliation(s)
- Santosh Narayan
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Gene Bryant
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shivangi Shah
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Georgina Berrozpe
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark Ptashne
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
11
|
Using c-kit to genetically target cerebellar molecular layer interneurons in adult mice. PLoS One 2017; 12:e0179347. [PMID: 28658323 PMCID: PMC5489153 DOI: 10.1371/journal.pone.0179347] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/26/2017] [Indexed: 11/19/2022] Open
Abstract
The cerebellar system helps modulate and fine-tune motor action. Purkinje cells (PCs) provide the sole output of the cerebellar cortex, therefore, any cerebellar involvement in motor activity must be driven by changes in PC firing rates. Several different cell types influence PC activity including excitatory input from parallel fibers and inhibition from molecular layer interneurons (MLIs). Similar to PCs, MLI activity is driven by parallel fibers, therefore, MLIs provide feed-forward inhibition onto PCs. To aid in the experimental assessment of how molecular layer inhibition contributes to cerebellar function and motor behavior, we characterized a new knock-in mouse line with Cre recombinase expression under control of endogenous c-kit transcriptional machinery. Using these engineered c-Kit mice, we were able to obtain high levels of conditional MLI transduction in adult mice using Cre-dependent viral vectors without any PC or granule cell labeling. We then used the mouse line to target MLIs for activity perturbation in vitro using opto- and chemogenetics.
Collapse
|
12
|
Lotinun S, Krishnamra N. Disruption of c-Kit Signaling in Kit(W-sh/W-sh) Growing Mice Increases Bone Turnover. Sci Rep 2016; 6:31515. [PMID: 27527615 PMCID: PMC4985756 DOI: 10.1038/srep31515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/21/2016] [Indexed: 12/13/2022] Open
Abstract
c-Kit tyrosine kinase receptor has been identified as a regulator of bone homeostasis. The c-Kit loss-of-function mutations in WBB6F1/J-KitW/W-v mice result in low bone mass. However, these mice are sterile and it is unclear whether the observed skeletal phenotype is secondary to a sex hormone deficiency. In contrast, C57BL/6J-KitW-sh/W-sh (Wsh/Wsh) mice, which carry an inversion mutation affecting the transcriptional regulatory elements of the c-Kit gene, are fertile. Here, we showed that Wsh/Wsh mice exhibited osteopenia with elevated bone resorption and bone formation at 6- and 9-week-old. The c-Kit Wsh mutation increased osteoclast differentiation, the number of committed osteoprogenitors, alkaline phosphatase activity and mineralization. c-Kit was expressed in both osteoclasts and osteoblasts, and c-Kit expression was decreased in Wsh/Wshosteoclasts, but not osteoblasts, suggesting an indirect effect of c-Kit on bone formation. Furthermore, the osteoclast-derived coupling factor Wnt10b mRNA was increased in Wsh/Wsh osteoclasts. Conditioned medium from Wsh/Wsh osteoclasts had elevated Wnt10b protein levels and induced increased alkaline phosphatase activity and mineralization in osteoblast cultures. Antagonizing Wnt10b signaling with DKK1 or Wnt10b antibody inhibited these effects. Our data suggest that c-Kit negatively regulates bone turnover, and disrupted c-Kit signaling couples increased bone resorption with bone formation through osteoclast-derived Wnt 10 b.
Collapse
Affiliation(s)
- Sutada Lotinun
- Department of Physiology and STAR on Craniofacial and Skeletal Disorders, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Nateetip Krishnamra
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Di Siena S, Gimmelli R, Nori SL, Barbagallo F, Campolo F, Dolci S, Rossi P, Venneri MA, Giannetta E, Gianfrilli D, Feigenbaum L, Lenzi A, Naro F, Cianflone E, Mancuso T, Torella D, Isidori AM, Pellegrini M. Activated c-Kit receptor in the heart promotes cardiac repair and regeneration after injury. Cell Death Dis 2016; 7:e2317. [PMID: 27468693 PMCID: PMC4973348 DOI: 10.1038/cddis.2016.205] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/01/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
The role of endogenous c-Kit receptor activation on cardiac cell homeostasis and repair remains largely unexplored. Transgenic mice carrying an activating point mutation (TgD814Y) in the kinase domain of the c-Kit gene were generated. c-KitTgD814Y receptor was expressed in the heart during embryonic development and postnatal life, in a similar timing and expression pattern to that of the endogenous gene, but not in the hematopoietic compartment allowing the study of a cardiac-specific phenotype. c-KitTgD814Y mutation produced a constitutive active c-Kit receptor in cardiac tissue and cells from transgenic mice as demonstrated by the increased phosphorylation of ERK1/2 and AKT, which are the main downstream molecular effectors of c-Kit receptor signaling. In adult transgenic hearts, cardiac morphology, size and total c-Kit+ cardiac cell number was not different compared with wt mice. However, when c-KitTgD814Y mice were subjected to transmural necrotic heart damage by cryoinjury (CI), all transgenic survived, compared with half of wt mice. In the sub-acute phase after CI, transgenic and wt mice showed similar heart damage. However, 9 days after CI, transgenic mice exhibited an increased number of c-Kit+CD31+ endothelial progenitor cells surrounding the necrotic area. At later follow-up, a consistent reduction of fibrotic area, increased capillary density and increased cardiomyocyte replenishment rate (as established by BrdU incorporation) were observed in transgenic compared with wt mice. Consistently, CD45−c-Kit+ cardiac stem cells isolated from transgenic c-KitTgD814Y mice showed an enhanced endothelial and cardiomyocyte differentiation potential compared with cells isolated from the wt. Constitutive activation of c-Kit receptor in mice is associated with an increased cardiac myogenic and vasculogenic reparative potential after injury, with a significant improvement of survival.
Collapse
Affiliation(s)
- S Di Siena
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - R Gimmelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S L Nori
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - F Barbagallo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Campolo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S Dolci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - P Rossi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - M A Venneri
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - L Feigenbaum
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer research, Frederick, MD, USA
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - E Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - T Mancuso
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - D Torella
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - M Pellegrini
- Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| |
Collapse
|
14
|
Fufa TD, Harris ML, Watkins-Chow DE, Levy D, Gorkin DU, Gildea DE, Song L, Safi A, Crawford GE, Sviderskaya EV, Bennett DC, Mccallion AS, Loftus SK, Pavan WJ. Genomic analysis reveals distinct mechanisms and functional classes of SOX10-regulated genes in melanocytes. Hum Mol Genet 2015; 24:5433-50. [PMID: 26206884 PMCID: PMC4572067 DOI: 10.1093/hmg/ddv267] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/09/2015] [Accepted: 07/06/2015] [Indexed: 12/31/2022] Open
Abstract
SOX10 is required for melanocyte development and maintenance, and has been linked to melanoma initiation and progression. However, the molecular mechanisms by which SOX10 guides the appropriate gene expression programs necessary to promote the melanocyte lineage are not fully understood. Here we employ genetic and epigenomic analysis approaches to uncover novel genomic targets and previously unappreciated molecular roles of SOX10 in melanocytes. Through global analysis of SOX10-binding sites and epigenetic characteristics of chromatin states, we uncover an extensive catalog of SOX10 targets genome-wide. Our findings reveal that SOX10 predominantly engages 'open' chromatin regions and binds to distal regulatory elements, including novel and previously known melanocyte enhancers. Integrated chromatin occupancy and transcriptome analysis suggest a role for SOX10 in both transcriptional activation and repression to regulate functionally distinct classes of genes. We demonstrate that distinct epigenetic signatures and cis-regulatory sequence motifs predicted to bind putative co-regulatory transcription factors define SOX10-activated and SOX10-repressed target genes. Collectively, these findings uncover a central role of SOX10 as a global regulator of gene expression in the melanocyte lineage by targeting diverse regulatory pathways.
Collapse
Affiliation(s)
- Temesgen D Fufa
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Melissa L Harris
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dawn E Watkins-Chow
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Denise Levy
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David U Gorkin
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Derek E Gildea
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lingyun Song
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA, Department of Pediatrics, Division of Molecular Genetics, Duke University, Durham, NC 27708, USA and
| | - Alexias Safi
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA, Department of Pediatrics, Division of Molecular Genetics, Duke University, Durham, NC 27708, USA and
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA, Department of Pediatrics, Division of Molecular Genetics, Duke University, Durham, NC 27708, USA and
| | - Elena V Sviderskaya
- Molecular Cell Sciences Research Centre, St George's, University of London, London SW17 0RE, UK
| | - Dorothy C Bennett
- Molecular Cell Sciences Research Centre, St George's, University of London, London SW17 0RE, UK
| | - Andrew S Mccallion
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA,
| |
Collapse
|
15
|
Molkentin JD. Letter by Molkentin regarding article, "The absence of evidence is not evidence of absence: the pitfalls of Cre Knock-Ins in the c-Kit Locus". Circ Res 2014; 115:e21-3. [PMID: 25258403 DOI: 10.1161/circresaha.114.305011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, OH
| |
Collapse
|
16
|
Nadal-Ginard B, Ellison GM, Torella D. Response to Molkentin’s Letter to The Editor Regarding Article, “The Absence of Evidence Is Not Evidence of Absence: The Pitfalls of Cre Knock-Ins in the c-kit Locus”. Circ Res 2014; 115:e38-9. [DOI: 10.1161/circresaha.115.305380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Bernardo Nadal-Ginard
- Center of Human and Aerospace Physiological Sciences and Center for Stem Cells and Regenerative Medicine, Faculty of Medicine & Life Sciences, King’s College London, London, United Kingdom
| | - Georgina M. Ellison
- Center of Human and Aerospace Physiological Sciences and Center for Stem Cells and Regenerative Medicine, Faculty of Medicine & Life Sciences, King’s College London, London, United Kingdom
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
17
|
Tong PL, Roediger B, Kolesnikoff N, Biro M, Tay SS, Jain R, Shaw LE, Grimbaldeston MA, Weninger W. The skin immune atlas: three-dimensional analysis of cutaneous leukocyte subsets by multiphoton microscopy. J Invest Dermatol 2014; 135:84-93. [PMID: 25007044 PMCID: PMC4268113 DOI: 10.1038/jid.2014.289] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/10/2014] [Accepted: 04/29/2014] [Indexed: 01/12/2023]
Abstract
Site-specific differences in skin response to pathogens and in the course of cutaneous inflammatory diseases are well appreciated. The composition and localization of cutaneous leukocytes has been studied extensively using histology and flow cytometry. However, the precise three-dimensional (3D) distribution of distinct immune cell subsets within skin at different body sites requires visualization of intact living skin. We used intravital multiphoton microscopy in transgenic reporter mice in combination with quantitative flow cytometry to generate a 3D immune cell atlas of mouse skin. The 3D location of innate and adaptive immune cells and site-specific differences in the densities of macrophages, T cells and mast cells at four defined sites (ear, back, footpad, tail) is presented. The combinatorial approach further demonstrates an as yet unreported age-dependent expansion of dermal gamma-delta T cells. Localization of dermal immune cells relative to anatomical structures was also determined. While dendritic cells were dispersed homogeneously within the dermis, mast cells preferentially localized to the perivascular space. Finally, we show the functional relevance of site-specific mast cell disparities using the passive cutaneous anaphylaxis model. These approaches are applicable to assessing immune cell variations and potential functional consequences in the setting of infection as well as the pathogenesis of inflammatory skin conditions.
Collapse
Affiliation(s)
- Philip L Tong
- Centenary Institute, Newtown, New South Wales, Australia; Discipline of Dermatology, The University of Sydney, Camperdown, New South Wales, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| | - Ben Roediger
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Natasha Kolesnikoff
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Maté Biro
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Szun S Tay
- Centenary Institute, Newtown, New South Wales, Australia
| | - Rohit Jain
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lisa E Shaw
- Centenary Institute, Newtown, New South Wales, Australia
| | | | - Wolfgang Weninger
- Centenary Institute, Newtown, New South Wales, Australia; Discipline of Dermatology, The University of Sydney, Camperdown, New South Wales, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| |
Collapse
|
18
|
David VA, Menotti-Raymond M, Wallace AC, Roelke M, Kehler J, Leighty R, Eizirik E, Hannah SS, Nelson G, Schäffer AA, Connelly CJ, O'Brien SJ, Ryugo DK. Endogenous retrovirus insertion in the KIT oncogene determines white and white spotting in domestic cats. G3 (BETHESDA, MD.) 2014; 4:1881-91. [PMID: 25085922 PMCID: PMC4199695 DOI: 10.1534/g3.114.013425] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/26/2014] [Indexed: 01/06/2023]
Abstract
The Dominant White locus (W) in the domestic cat demonstrates pleiotropic effects exhibiting complete penetrance for absence of coat pigmentation and incomplete penetrance for deafness and iris hypopigmentation. We performed linkage analysis using a pedigree segregating White to identify KIT (Chr. B1) as the feline W locus. Segregation and sequence analysis of the KIT gene in two pedigrees (P1 and P2) revealed the remarkable retrotransposition and evolution of a feline endogenous retrovirus (FERV1) as responsible for two distinct phenotypes of the W locus, Dominant White, and white spotting. A full-length (7125 bp) FERV1 element is associated with white spotting, whereas a FERV1 long terminal repeat (LTR) is associated with all Dominant White individuals. For purposes of statistical analysis, the alternatives of wild-type sequence, FERV1 element, and LTR-only define a triallelic marker. Taking into account pedigree relationships, deafness is genetically linked and associated with this marker; estimated P values for association are in the range of 0.007 to 0.10. The retrotransposition interrupts a DNAase I hypersensitive site in KIT intron 1 that is highly conserved across mammals and was previously demonstrated to regulate temporal and tissue-specific expression of KIT in murine hematopoietic and melanocytic cells. A large-population genetic survey of cats (n = 270), representing 30 cat breeds, supports our findings and demonstrates statistical significance of the FERV1 LTR and full-length element with Dominant White/blue iris (P < 0.0001) and white spotting (P < 0.0001), respectively.
Collapse
Affiliation(s)
- Victor A David
- Laboratory of Genomic Diversity, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Marilyn Menotti-Raymond
- Laboratory of Genomic Diversity, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Andrea Coots Wallace
- Laboratory of Genomic Diversity, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702
| | - Melody Roelke
- Leidos Biomedical Research Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702 Labooratory Animal Sciences Program (LASP) Bethesda Leidos Biomedical Research, Bethesda, Maryland 20892-2471
| | - James Kehler
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814
| | - Robert Leighty
- Data Management Services, Inc., National Cancer Institute-Frederick, Frederick, Maryland 21702
| | - Eduardo Eizirik
- Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90619-900, Brazil Instituto Pró-Carnívoros, Atibaia, Sao Paulo 12945-010, Brazil
| | | | - George Nelson
- BSP-CCR Genetics Core, Frederick National Laboratory, Frederick, Maryland 21702
| | - Alejandro A Schäffer
- National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland 20894
| | | | - Stephen J O'Brien
- Laboratory of Genomic Diversity, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702 Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
| | - David K Ryugo
- Department of Otolaryngology, Head and Neck Surgery, Center for Hearing Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Nadal-Ginard B, Ellison GM, Torella D. Absence of evidence is not evidence of absence: pitfalls of cre knock-ins in the c-Kit locus. Circ Res 2014; 115:415-8. [PMID: 24965482 DOI: 10.1161/circresaha.114.304676] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bernardo Nadal-Ginard
- From the Centre of Human and Aerospace Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, King's College London, London, United Kingdom (B.N.-G., G.M.E.); and Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy (G.M.E., D.T.).
| | - Georgina M Ellison
- From the Centre of Human and Aerospace Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, King's College London, London, United Kingdom (B.N.-G., G.M.E.); and Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy (G.M.E., D.T.)
| | - Daniele Torella
- From the Centre of Human and Aerospace Physiological Sciences and Centre for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, King's College London, London, United Kingdom (B.N.-G., G.M.E.); and Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy (G.M.E., D.T.).
| |
Collapse
|
20
|
Berrozpe G, Bryant GO, Warpinski K, Ptashne M. Regulation of a mammalian gene bearing a CpG island promoter and a distal enhancer. Cell Rep 2013; 4:445-53. [PMID: 23911283 DOI: 10.1016/j.celrep.2013.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/04/2013] [Accepted: 07/01/2013] [Indexed: 11/30/2022] Open
Abstract
A quantitative nucleosome occupancy assay revealed rules for nucleosome disposition in yeast and showed how disposition affects regulation of the GAL genes. Here, we show how those findings apply to the control of Kit, a mammalian gene. The Kit promoter lies in a CpG island, and its enhancer (active in mast cells) lies some 150 kb upstream. Nucleosomes form with especially high avidities at the Kit promoter, a reaction that, we surmise, ensures extremely low basal expression. In mast cells, transcriptional activators displace nucleosomes that are less tightly formed at the Kit enhancer. In turn, the active enhancer replaces a single Kit promoter nucleosome with the transcriptional machinery, thereby inducing transcription over 1,000-fold. As at the yeast GAL genes, the inhibitory effects of nucleosomes facilitate high factors of induction by mammalian activators working in the absence of specific repressors.
Collapse
Affiliation(s)
- Georgina Berrozpe
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, 430 East 67(th) Street, New York, NY 10065, USA
| | | | | | | |
Collapse
|
21
|
Michel A, Schüler A, Friedrich P, Döner F, Bopp T, Radsak M, Hoffmann M, Relle M, Distler U, Kuharev J, Tenzer S, Feyerabend TB, Rodewald HR, Schild H, Schmitt E, Becker M, Stassen M. Mast cell-deficient Kit(W-sh) "Sash" mutant mice display aberrant myelopoiesis leading to the accumulation of splenocytes that act as myeloid-derived suppressor cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:5534-44. [PMID: 23636054 DOI: 10.4049/jimmunol.1203355] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mast cell-deficient Kit(W-sh) "sash" mice are widely used to investigate mast cell functions. However, mutations of c-Kit also affect additional cells of hematopoietic and nonimmune origin. In this study, we demonstrate that Kit(W-sh) causes aberrant extramedullary myelopoiesis characterized by the expansion of immature lineage-negative cells, common myeloid progenitors, and granulocyte/macrophage progenitors in the spleen. A consistent feature shared by these cell types is the reduced expression of c-Kit. Populations expressing intermediate and high levels of Ly6G, a component of the myeloid differentiation Ag Gr-1, are also highly expanded in the spleen of sash mice. These cells are able to suppress T cell responses in vitro and phenotypically and functionally resemble myeloid-derived suppressor cells (MDSC). MDSC typically accumulate in tumor-bearing hosts and are able to dampen immune responses. Consequently, transfer of MDSC from naive sash mice into line 1 alveolar cell carcinoma tumor-bearing wild-type littermates leads to enhanced tumor progression. However, although it can also be observed in sash mice, accelerated growth of transplanted line 1 alveolar cell carcinoma tumors is a mast cell-independent phenomenon. Thus, the Kit(W-sh) mutation broadly affects key steps in myelopoiesis that may have an impact on mast cell research.
Collapse
Affiliation(s)
- Anastasija Michel
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Roediger B, Kyle R, Yip KH, Sumaria N, Guy TV, Kim BS, Mitchell AJ, Tay SS, Jain R, Forbes-Blom E, Chen X, Tong PL, Bolton HA, Artis D, Paul WE, Fazekas de St Groth B, Grimbaldeston MA, Le Gros G, Weninger W. Cutaneous immunosurveillance and regulation of inflammation by group 2 innate lymphoid cells. Nat Immunol 2013; 14:564-73. [PMID: 23603794 DOI: 10.1038/ni.2584] [Citation(s) in RCA: 385] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/05/2013] [Indexed: 02/06/2023]
Abstract
Type 2 immunity is critical for defense against cutaneous infections but also underlies the development of allergic skin diseases. We report the identification in normal mouse dermis of an abundant, phenotypically unique group 2 innate lymphoid cell (ILC2) subset that depended on interleukin 7 (IL-7) and constitutively produced IL-13. Intravital multiphoton microscopy showed that dermal ILC2 cells specifically interacted with mast cells, whose function was suppressed by IL-13. Treatment of mice deficient in recombination-activating gene 1 (Rag1(-/-)) with IL-2 resulted in the population expansion of activated, IL-5-producing dermal ILC2 cells, which led to spontaneous dermatitis characterized by eosinophil infiltrates and activated mast cells. Our data show that ILC2 cells have both pro- and anti-inflammatory properties and identify a previously unknown interactive pathway between two innate populations of cells of the immune system linked to type 2 immunity and allergic diseases.
Collapse
|
23
|
Rodewald HR, Feyerabend TB. Widespread immunological functions of mast cells: fact or fiction? Immunity 2012; 37:13-24. [PMID: 22840840 DOI: 10.1016/j.immuni.2012.07.007] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Indexed: 12/29/2022]
Abstract
Immunological functions of mast cells are currently considered to be much broader than the original role of mast cells in IgE-driven allergic disease. The spectrum of proposed mast cell functions includes areas as diverse as the regulation of innate and adaptive immune responses, protective immunity against viral, microbial, and parasitic pathogens, autoimmunity, tolerance to graft rejection, promotion of or protection from cancer, wound healing, angiogenesis, cardiovascular diseases, diabetes, obesity, and others. The vast majority of in vivo mast cell data have been based on mast cell-deficient Kit mutant mice. However, work in new mouse mutants with unperturbed Kit function, which have a surprisingly normal immune system, has failed to corroborate some key immunological aspects, formerly attributed to mast cells. Here, we consider the implications of these recent developments for the state of the field as well as for future work, aiming at deciphering the physiological functions of mast cells.
Collapse
Affiliation(s)
- Hans-Reimer Rodewald
- Division for Cellular Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany.
| | | |
Collapse
|
24
|
Becker M, Reuter S, Friedrich P, Doener F, Michel A, Bopp T, Klein M, Schmitt E, Schild H, Radsak MP, Echtenacher B, Taube C, Stassen M. Genetic variation determines mast cell functions in experimental asthma. THE JOURNAL OF IMMUNOLOGY 2011; 186:7225-31. [PMID: 21572035 DOI: 10.4049/jimmunol.1100676] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mast cell-deficient mice are a key for investigating the function of mast cells in health and disease. Allergic airway disease induced as a Th2-type immune response in mice is employed as a model to unravel the mechanisms underlying inception and progression of human allergic asthma. Previous work done in mast cell-deficient mouse strains that otherwise typically mount Th1-dominated immune responses revealed contradictory results as to whether mast cells contribute to the development of airway hyperresponsiveness and airway inflammation. However, a major contribution of mast cells was shown using adjuvant-free protocols to achieve sensitization. The identification of a traceable genetic polymorphism closely linked to the Kit(W-sh) allele allowed us to generate congenic mast cell-deficient mice on a Th2-prone BALB/c background, termed C.B6-Kit(W-sh). In accordance with the expectations, C.B6-Kit(W-sh) mice do not develop IgE- and mast cell-dependent passive cutaneous anaphylaxis. Yet, unexpectedly, C.B6-Kit(W-sh) mice develop full-blown airway inflammation, airway hyperresponsiveness, and mucus production despite the absence of mast cells. Thus, our findings demonstrate a major influence of genetic background on the contribution of mast cells in an important disease model and introduce a novel strain of mast cell-deficient mice.
Collapse
Affiliation(s)
- Marc Becker
- Institute for Immunology, Medical Center, Johannes Gutenberg University, Mainz D-55131, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mast cell hyperplasia, B-cell malignancy, and intestinal inflammation in mice with conditional expression of a constitutively active kit. Blood 2010; 117:2012-21. [PMID: 21148330 DOI: 10.1182/blood-2008-11-189605] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Signaling through the receptor tyrosine kinase kit controls proliferation and differentiation of hematopoietic precursor cells and mast cells. Somatic point mutations of the receptor that constitutively activate kit signaling are associated with mastocytosis and various hematopoietic malignancies. We generated a Cre/loxP-based bacterial artificial chromosome transgenic mouse model that allows conditional expression of a kit gene carrying the kitD814V mutation (the murine homolog of the most common mutation in human mastocytosis, kitD816V) driven by the kit promoter. Expression of the mutant kit in cells of adult mice, including hematopoietic precursors, caused severe mastocytosis with 100% penetrance at young age frequently associated with additional hematopoietic (mostly B lineage-derived) neoplasms and focal colitis. Restriction of transgene expression to mature mast cells resulted in a similar mast cell disease developing with slower kinetics. Embryonic expression led to a hyperproliferative dysregulation of the erythroid lineage with a high rate of perinatal lethality. In addition, most adult animals developed colitis associated with mucosal mast cell accumulation. Our findings demonstrate that the effects of constitutive kit signaling critically depend on the developmental stage and the state of differentiation of the cell hit by the gain-of-function mutation.
Collapse
|
26
|
Ro S, Park C, Jin J, Zheng H, Blair PJ, Redelman D, Ward SM, Yan W, Sanders KM. A model to study the phenotypic changes of interstitial cells of Cajal in gastrointestinal diseases. Gastroenterology 2010; 138:1068-78.e782. [PMID: 19917283 PMCID: PMC4793910 DOI: 10.1053/j.gastro.2009.11.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/27/2009] [Accepted: 11/05/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Interstitial cells of Cajal (ICC) express the receptor tyrosine kinase, KIT, the receptor for stem cell factor. In the gastrointestinal (GI) tract, ICC are pacemaker cells that generate spontaneous electrical slow waves, and mediate inputs from motor neurons. Absence or loss of ICC are associated with GI motility disorders, including those consequent of diabetes. Studies of ICC have been hampered by the low density of these cells and difficulties in recognizing these cells in cell dispersions. METHODS Kit(+/copGFP) mice harboring a copepod super green fluorescent protein (copGFP) complementary DNA, inserted at the Kit locus, were generated. copGFP(+) ICC from GI muscles were analyzed using confocal microscopy and flow cytometry. copGFP(+) ICC from the jejunum were purified by a fluorescence-activated cell sorter and validated by cell-specific markers. Kit(+/copGFP) mice were crossbred with diabetic Lep(+/ob) mice to generate compound Kit(+/copGFP);Lep(ob/ob) mutant mice. copGFP(+) ICC from compound transgenic mice were analyzed by confocal microscopy. RESULTS copGFP in Kit(+/copGFP) mice colocalized with KIT immunofluorescence and thus was predominantly found in ICC. In other smooth muscles, mast cells were also labeled, but these cells were relatively rare in the murine GI tract. copGFP(+) cells from jejunal muscles were Kit(+) and free of contaminating cell-specific markers. Kit(+/copGFP);Lep(ob/ob) mice displayed ICC networks that were dramatically disrupted during the development of diabetes. CONCLUSIONS Kit(+/copGFP) mice offer a powerful new model to study the function and genetic regulation of ICC phenotypes. Isolation of ICC from animal models will help determine the causes and responses of ICC to therapeutic agents.
Collapse
Affiliation(s)
- Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Jingling Jin
- Huffington Center on Aging and Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Peter J. Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Doug Redelman
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
27
|
Itoh S, Nakae S, Velotta JB, Kosuge H, Connolly A, Tsai M, Adachi H, Galli SJ, Robbins RC, Fischbein MP. The role of recipient mast cells in acute and chronic cardiac allograft rejection in C57BL/6-KitW-sh/W-sh mice. J Heart Lung Transplant 2009; 29:401-9. [PMID: 19818646 DOI: 10.1016/j.healun.2009.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 08/18/2009] [Accepted: 08/22/2009] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Mast cells are hypothesized to promote rejection and adverse remodeling in cardiac allografts. In contrast, it has been reported that mast cells may enhance cardiac allograft survival in rats. We used C57BL/6-Kit(W-sh/W-sh) mast cell-deficient and corresponding wild-type mice to investigate possible contributions of recipient mast cells to acute or chronic cardiac allograft rejection. METHODS FVB (H-2(q); acute rejection), or C-H-2(bm12)KhEg (H-2(bm12); chronic rejection) donor hearts were heterotopically transplanted into C57BL/6-Kit(W-sh/W-sh) (H-2(b)) or C57BL/6-Kit(+/+) (H-2(b)) mice. The degree of acute rejection was assessed at 5 days and chronic rejection, at 52 days. RESULTS In the acute rejection model, donor heart vascular cell adhesion molecule-1 (VCAM-1) expression was significantly lower in C57BL/6-Kit(W-sh/W-sh) than in wild-type recipients; however, acute rejection scores, graft survival, inflammatory cells, or cytokine expression did not differ significantly. In the chronic rejection model, the number of mast cells/mm(2) of allograft tissue was significantly increased 52 days after transplantation in allografts transplanted into C57BL/6-Kit(+/+) but not C57BL/6-Kit(W-sh/W-sh) mice; however, no substantial differences were noted in graft coronary artery disease, graft inflammatory cells, or levels of graft tissue expression of cytokines or adhesion molecules. CONCLUSIONS Cardiac allografts undergoing chronic rejection in wild-type C57BL/6-Kit(+/+) mice exhibit increased numbers of mast cells, but acute or chronic cardiac allograft rejection can develop in C57BL/6-Kit(W-sh/W-sh) mice even though these recipients virtually lack mast cells. These findings indicate that recipient mast cells are not required for acute or chronic cardiac allograft rejection in the models examined.
Collapse
Affiliation(s)
- Satoshi Itoh
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mithraprabhu S, Loveland KL. Control of KIT signalling in male germ cells: what can we learn from other systems? Reproduction 2009; 138:743-57. [PMID: 19567460 DOI: 10.1530/rep-08-0537] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The KIT ligand (KITL)/KIT-signalling system is among several pathways known to be essential for fertility. In the postnatal testis, the KIT/KITL interaction is crucial for spermatogonial proliferation, differentiation, survival and subsequent entry into meiosis. Hence, identification of endogenous factors that regulate KIT synthesis is important for understanding the triggers driving germ cell maturation. Although limited information is available regarding local factors in the testicular microenvironment that modulate KIT synthesis at the onset of spermatogenesis, knowledge from other systems could be used as a basis for identifying how KIT function is regulated in germ cells. This review describes the known regulators of KIT, including transcription factors implicated in KIT promoter regulation. In addition, specific downstream outcomes in biological processes that KIT orchestrates are addressed. These are discussed in relationship to current knowledge of mammalian germ cell development.
Collapse
Affiliation(s)
- Sridurga Mithraprabhu
- Monash Institute for Medical Research, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
29
|
Cerisoli F, Cassinelli L, Lamorte G, Citterio S, Bertolotti F, Magli MC, Ottolenghi S. Green fluorescent protein transgene driven by Kit regulatory sequences is expressed in hematopoietic stem cells. Haematologica 2009; 94:318-25. [PMID: 19181779 DOI: 10.3324/haematol.13689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The transcriptional regulation of stem cell genes is still poorly understood. Kit, encoding the stem cell factor receptor, is a pivotal molecule for multiple types of stem/progenitor cells. We previously generated mouse lines expressing transgenic green fluorescent protein under the control of Kit promoter/first intron regulatory elements, and we demonstrated expression in hematopoietic progenitors. DESIGN AND METHODS In the present work we investigated whether the transgene is also expressed in hematopoietic stem cells of adult bone marrow and fetal liver. To this purpose, we tested, in long-term repopulating assays, cell fractions expressing different levels of green fluorescent protein within Kit-positive or SLAM-selected populations. RESULTS The experiments demonstrated transgene expression in both fetal and adult hematopoietic stem cells and indicated that the transgene is transcribed at distinctly lower levels in hematopoietic stem cells than in pluripotent and committed progenitors. CONCLUSIONS These results, together with previous data, show that a limited subset of DNA sequences drives gene expression in number of stem cell types (hematopoietic stem cells, primordial germ cells, cardiac stem cells). Additionally, our results might help to further improve high level purification of hematopoietic stem cells for experimental purposes. Finally, as the Kit/green fluorescent protein transgene is expressed in multiple stem cell types, our transgenic model provides powerful in vivo system to track these cells during development and tissue regeneration.
Collapse
Affiliation(s)
- Francesco Cerisoli
- Institute of Biomedical Technologies, National Council of Research, Pisa, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Nigrovic PA, Gray DHD, Jones T, Hallgren J, Kuo FC, Chaletzky B, Gurish M, Mathis D, Benoist C, Lee DM. Genetic inversion in mast cell-deficient (Wsh) mice interrupts corin and manifests as hematopoietic and cardiac aberrancy. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1693-701. [PMID: 18988802 DOI: 10.2353/ajpath.2008.080407] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mast cells participate in pathophysiological processes that range from antimicrobial defense to anaphylaxis and inflammatory arthritis. Much of the groundwork for the understanding of mast cells was established in mice that lacked mast cells through defects in either stem cell factor or its receptor, Kit. Among available strains, C57BL/6-Kit(W-sh) (W(sh)) mice are experimentally advantageous because of their background strain and fertility. However, the genetic inversion responsible for the W(sh) phenotype remains poorly defined, and its effects beyond the mast cell have been incompletely characterized. We report that W(sh) animals exhibit splenomegaly with expanded myeloid and megakaryocyte populations. Hematopoietic abnormalities extend to the bone marrow and are reflected by neutrophilia and thrombocytosis. In contrast, mast cell-deficient WBB6F1-Kit(W)/Kit(W-v) (W/W(v)) mice display mild neutropenia, but no changes in circulating platelet numbers. To help define the basis for the W(sh) phenotype, a "DNA walking" strategy was used to identify the precise location of the 3' breakpoint, which was found to reside 67.5 kb upstream of Kit. The 5' breakpoint disrupts corin, a cardiac protease responsible for the activation of atrial natriuretic peptide. Consistent with this result, transcription of full-length corin is ablated and W(sh) mice develop symptoms of cardiomegaly. Studies performed using mast cell-deficient strains must consider the capacity of associated abnormalities to either expose or compensate for the missing mast cell lineage.
Collapse
Affiliation(s)
- Peter A Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jing H, Vakoc CR, Ying L, Mandat S, Wang H, Zheng X, Blobel GA. Exchange of GATA factors mediates transitions in looped chromatin organization at a developmentally regulated gene locus. Mol Cell 2008; 29:232-42. [PMID: 18243117 PMCID: PMC2254447 DOI: 10.1016/j.molcel.2007.11.020] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 09/21/2007] [Accepted: 11/09/2007] [Indexed: 12/18/2022]
Abstract
Enhancers can regulate designate promoters over long distances by forming chromatin loops. Whether chromatin loops are lost or reconfigured during gene repression is largely unexplored. We examined the chromosome conformation of the Kit gene that is expressed during early erythropoiesis but is downregulated upon cell maturation. Kit expression is controlled by sequential occupancy of two GATA family transcription factors. In immature cells, a distal enhancer bound by GATA-2 is in physical proximity with the active Kit promoter. Upon cell maturation, GATA-1 displaces GATA-2 and triggers a loss of the enhancer/promoter interaction. Moreover, GATA-1 reciprocally increases the proximity in nuclear space among distinct downstream GATA elements. GATA-1-induced transitions in chromatin conformation are not simply the consequence of transcription inhibition and require the cofactor FOG-1. This work shows that a GATA factor exchange reconfigures higher-order chromatin organization, and suggests that de novo chromatin loop formation is employed by nuclear factors to specify repressive outcomes.
Collapse
Affiliation(s)
- Huie Jing
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Nebert DW, Zhang G, Vesell ES. From human genetics and genomics to pharmacogenetics and pharmacogenomics: past lessons, future directions. Drug Metab Rev 2008; 40:187-224. [PMID: 18464043 PMCID: PMC2752627 DOI: 10.1080/03602530801952864] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A brief history of human genetics and genomics is provided, comparing recent progress in those fields with that in pharmacogenetics and pharmacogenomics, which are subsets of genetics and genomics, respectively. Sequencing of the entire human genome, the mapping of common haplotypes of single-nucleotide polymorphisms (SNPs), and cost-effective genotyping technologies leading to genome-wide association (GWA) studies - have combined convincingly in the past several years to demonstrate the requirements needed to separate true associations from the plethora of false positives. While research in human genetics has moved from monogenic to oligogenic to complex diseases, its pharmacogenetics branch has followed, usually a few years behind. The continuous discoveries, even today, of new surprises about our genome cause us to question reviews declaring that "personalized medicine is almost here" or that "individualized drug therapy will soon be a reality." As summarized herein, numerous reasons exist to show that an "unequivocal genotype" or even an "unequivocal phenotype" is virtually impossible to achieve in current limited-size studies of human populations. This problem (of insufficiently stringent criteria) leads to a decrease in statistical power and, consequently, equivocal interpretation of most genotype-phenotype association studies. It remains unclear whether personalized medicine or individualized drug therapy will ever be achievable by means of DNA testing alone.
Collapse
Affiliation(s)
- Daniel W Nebert
- Division of Human Genetics, Department of Pediatrics & Molecular Developmental Biology, Cincinnati, Ohio 45267-0056, USA.
| | | | | |
Collapse
|
33
|
Zhou JS, Xing W, Friend DS, Austen KF, Katz HR. Mast cell deficiency in Kit(W-sh) mice does not impair antibody-mediated arthritis. ACTA ACUST UNITED AC 2007; 204:2797-802. [PMID: 17998392 PMCID: PMC2118523 DOI: 10.1084/jem.20071391] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
We previously reported that joint swelling, synovial thickening, and cartilage matrix depletion induced by the injection of anti-collagen monoclonal antibodies and lipopolysaccharide (LPS) in BALB/c mice are increased in the absence of inhibitory leukocyte immunoglobulin (Ig)-like receptor B4 (LILRB4; formerly gp49B1) in a neutrophil-dependent manner. Because both mast cells and neutrophils express LILRB4, we sought a mast cell requirement with mast cell-deficient mouse strains, but unexpectedly obtained full arthritis in Kit(W-sh) mice and full resistance in Kit(W/KitW-v) mice. Kit(W-sh) mice were indeed mast cell deficient as assessed by histology and the absence of IgE/mast cell-dependent passive cutaneous anaphylaxis in the ear and joint as well as passive systemic anaphylaxis. Deletion of LILRB4 in Kit(W-sh) mice exacerbated anti-collagen/LPS-induced joint swelling that was abolished by neutrophil depletion, establishing a counterregulatory role for LILRB4 in the absence of mast cells. Whereas blood neutrophil levels and LPS-elicited tissue neutrophilia were equal in Kit(W-sh) and Kit+ mice, both were impaired in Kit(W/KitW-v) mice. Although both strains are mast cell deficient and protected from IgE-mediated anaphylactic reactions, their dramatically different responses to autoantibody-mediated, neutrophil-dependent immune complex arthritis suggest that other host differences determine the extent of mast cell involvement. Thus, a conclusion for an absolute mast cell role in a pathobiologic process requires evidence from both strains.
Collapse
Affiliation(s)
- Joseph S Zhou
- Department of Medicine, Harvard Medical School, and Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
34
|
Filipponi D, Hobbs RM, Ottolenghi S, Rossi P, Jannini EA, Pandolfi PP, Dolci S. Repression of kit expression by Plzf in germ cells. Mol Cell Biol 2007; 27:6770-81. [PMID: 17664282 PMCID: PMC2099235 DOI: 10.1128/mcb.00479-07] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Male mice lacking expression of Plzf, a DNA sequence-specific transcriptional repressor, show progressive germ cell depletion due to exhaustion of the spermatogonial stem cell population. This is likely due to the deregulated expression of genes controlling the switch between spermatogonial self-renewal and differentiation. Here we show that Plzf directly represses the transcription of kit, a hallmark of spermatogonial differentiation. Plzf represses both endogenous kit expression and expression of a reporter gene under the control of the kit promoter region. A discrete sequence of the kit promoter, required for Plzf-mediated kit transcriptional repression, is bound by Plzf both in vivo and in vitro. A 3-bp mutation in this Plzf binding site abolishes the responsiveness of the kit promoter to Plzf repression. A significant increase in kit expression is also found in the undifferentiated spermatogonia isolated from Plzf(-/-) mice. Thus, we suggest that one mechanism by which Plzf maintains the pool of spermatogonial stem cells is through a direct repression of kit expression.
Collapse
Affiliation(s)
- Doria Filipponi
- Department of Public Health and Cell Biology, University of Rome Tor Vergata, Via Montpellier 1, Ed E Nord, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Nigrovic PA, Binstadt BA, Monach PA, Johnsen A, Gurish M, Iwakura Y, Benoist C, Mathis D, Lee DM. Mast cells contribute to initiation of autoantibody-mediated arthritis via IL-1. Proc Natl Acad Sci U S A 2007; 104:2325-30. [PMID: 17277081 PMCID: PMC1892913 DOI: 10.1073/pnas.0610852103] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mast cells are immune sentinels that participate in the defense against bacteria and parasites. Resident within the joint, mast cells become activated in human rheumatoid arthritis and are implicated in the pathogenesis of experimental murine synovitis. However, their arthritogenic role remains undefined. Using a model of autoantibody-induced arthritis, we show that mast cells contribute to the initiation of inflammation within the joint by elaboration of IL-1. Mast cells become activated to produce this cytokine via the IgG immune complex receptor FcgammaRIII. Interestingly, mast cells become dispensable for the perpetuation of arthritis after delivery of IL-1, highlighting the contribution of this lineage to arthritis induction. These findings illuminate a mechanism by which mast cells can participate in the pathogenesis of autoimmune inflammatory arthritis and provide insights of potential relevance to human rheumatoid arthritis.
Collapse
Affiliation(s)
- Peter A. Nigrovic
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- Division of Immunology, Children's Hospital Boston, Boston, MA 02115
| | - Bryce A. Binstadt
- Division of Immunology, Children's Hospital Boston, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215; and
| | - Paul A. Monach
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215; and
| | - Alyssa Johnsen
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215; and
| | - Michael Gurish
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
| | - Yoichiro Iwakura
- Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Christophe Benoist
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215; and
| | - Diane Mathis
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215; and
- To whom correspondence may be addressed at:
Section on Immunology and Immunogenetics, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215. E-mail:
| | - David M. Lee
- *Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115
- To whom correspondence may be addressed at:
Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, One Jimmy Fund Way, Smith 552B, Boston, MA 02115. E-mail:
| |
Collapse
|