1
|
Caviness PC, Lazarenko OP, Blackburn ML, Chen JR. Sex-dependent effect of GPR109A gene deletion in myeloid cells on bone development in mice. RESEARCH SQUARE 2025:rs.3.rs-6206075. [PMID: 40235504 PMCID: PMC11998753 DOI: 10.21203/rs.3.rs-6206075/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Blueberry metabolite-derived phenolic acids are thought to suppress bone resorption via interactions with the G protein-coupled receptor 109A (GPR109A). Previously, global GPR109A knockout (GPR109A ⁻/⁻ ) mice exhibited increased bone mass and a diminished bone-protective response to phenolic acids. While GPR109A is highly expressed in osteoclast precursor macrophages, its role in bone development remains unclear. To address this, we generated a myeloid cell-specific GPR109A knockout (GPR109A flox/flox /LysM-Cre⁺; CKO) mouse model and assessed bone phenotypes in male and female mice at 35 days, 3 months, 6 months, and 12 months using µCT. At 35 days, CKO males showed significantly improved tibia and vertebrae µCT parameters compared to controls (f/f, Cre⁺). However, at later time points (6 and 12 months), Cre recombinase effects were observed, with Cre⁺ males exhibiting similar bone parameters to CKO mice. In contrast, female CKO mice displayed significantly improved µCT parameters at 6 and 12 months. Notably, 12-month-old Cre⁺ males exhibited altered bone mechanical properties, while females did not. Gene expression analysis revealed increased Interferon regulatory factor 8 (Irf8), an osteoclastogenesis suppressor, in female CKO mice. These findings suggest that GPR109A regulates bone resorption through osteoclastogenic pathways in a sex-specific manner.
Collapse
|
2
|
Gimenez GA, Romijn M, van den Herik J, Meijer W, Eggers R, Hobo B, De Zeeuw CI, Canto CB, Verhaagen J, Carulli D. A Study on Potential Sources of Perineuronal Net-Associated Sema3A in Cerebellar Nuclei Reveals Toxicity of Non-Invasive AAV-Mediated Cre Expression in the Central Nervous System. Int J Mol Sci 2025; 26:819. [PMID: 39859534 PMCID: PMC11765860 DOI: 10.3390/ijms26020819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Semaphorin 3A (Sema3A) is an axon guidance molecule, which is also abundant in the adult central nervous system (CNS), particularly in perineuronal nets (PNNs). PNNs are extracellular matrix structures that restrict plasticity. The cellular sources of Sema3A in PNNs are unknown. Most Sema3A-bearing neurons do not express Sema3A mRNA, suggesting that Sema3A may be released from other neurons. Another potential source of Sema3A is the choroid plexus. To identify sources of PNN-associated Sema3A, we focused on the cerebellar nuclei, which contain Sema3A+ PNNs. Cerebellar nuclei neurons receive prominent input from Purkinje cells (PCs), which express high levels of Sema3A mRNA. By using a non-invasive viral vector approach, we overexpressed Cre in PCs, the choroid plexus, or throughout the CNS of Sema3Afl/fl mice. Knocking out Sema3A in PCs or the choroid plexus was not sufficient to decrease the amount of PNN-associated Sema3A. Alternatively, knocking out Sema3A throughout the CNS induced a decrease in PNN-associated Sema3A. However, motor deficits, microgliosis, and neurodegeneration were observed, which were due to Cre toxicity. Our study represents the first attempt to unravel cellular sources of PNN-associated Sema3A and shows that non-invasive viral-mediated Cre expression throughout the CNS could lead to toxicity, complicating the interpretation of Cre-mediated Sema3A knock-out.
Collapse
Affiliation(s)
- Geoffrey-Alexander Gimenez
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Maurits Romijn
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Joëlle van den Herik
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Wouter Meijer
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Barbara Hobo
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| | - Chris I. De Zeeuw
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Cathrin B. Canto
- Department of Cerebellar Coordination & Cognition, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (C.I.D.Z.); (C.B.C.)
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Daniela Carulli
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands; (G.-A.G.); (M.R.); (J.v.d.H.); (W.M.); (R.E.); (B.H.); (J.V.)
| |
Collapse
|
3
|
Lyu QR, Fu K. Tissue-specific Cre driver mice to study vascular diseases. Vascul Pharmacol 2023; 153:107241. [PMID: 37923099 DOI: 10.1016/j.vph.2023.107241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Vascular diseases, including atherosclerosis and abdominal aneurysms, are the primary cause of mortality and morbidity among the elderly worldwide. The life quality of patients is significantly compromised due to inadequate therapeutic approaches and limited drug targets. To expand our comprehension of vascular diseases, gene knockout (KO) mice, especially conditional knockout (cKO) mice, are widely used for investigating gene function and mechanisms of action. The Cre-loxP system is the most common method for generating cKO mice. Numerous Cre driver mice have been established to study the main cell types that compose blood vessels, including endothelial cells, smooth muscle cells, and fibroblasts. Here, we first discuss the characteristics of each layer of the arterial wall. Next, we provide an overview of the representative Cre driver mice utilized for each of the major cell types in the vessel wall and their most recent applications in vascular biology. We then go over Cre toxicity and discuss the practical methods for minimizing Cre interference in experimental outcomes. Finally, we look into the future of tissue-specific Cre drivers by introducing the revolutionary single-cell RNA sequencing and dual recombinase system.
Collapse
Affiliation(s)
- Qing Rex Lyu
- Medical Research Center, Chongqing General Hospital, Chongqing 401147, China; Chongqing Academy of Medical Sciences, Chongqing 401147, China.
| | - Kailong Fu
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
4
|
Cunningham CJ, Choi RB, Bullock WA, Robling AG. Perspective: The current state of Cre driver mouse lines in skeletal research: Challenges and opportunities. Bone 2023; 170:116719. [PMID: 36868507 PMCID: PMC10087282 DOI: 10.1016/j.bone.2023.116719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
The Cre/Lox system has revolutionized the ability of biomedical researchers to ask very specific questions about the function of individual genes in specific cell types at specific times during development and/or disease progression in a variety of animal models. This is true in the skeletal biology field, and numerous Cre driver lines have been created to foster conditional gene manipulation in specific subpopulations of bone cells. However, as our ability to scrutinize these models increases, an increasing number of issues have been identified with most driver lines. All existing skeletal Cre mouse models exhibit problems in one or more of the following three areas: (1) cell type specificity-avoiding Cre expression in unintended cell types; (2) Cre inducibility-improving the dynamic range for Cre in inducible models (negligible Cre activity before induction and high Cre activity after induction); and (3) Cre toxicity-reducing the unwanted biological effects of Cre (beyond loxP recombination) on cellular processes and tissue health. These issues are hampering progress in understanding the biology of skeletal disease and aging, and consequently, identification of reliable therapeutic opportunities. Skeletal Cre models have not advanced technologically in decades despite the availability of improved tools, including multi-promoter-driven expression of permissive or fragmented recombinases, new dimerization systems, and alternative forms of recombinases and DNA sequence targets. We review the current state of skeletal Cre driver lines, and highlight some of the successes, failures, and opportunities to improve fidelity in the skeleton, based on successes pioneered in other areas of biomedical science.
Collapse
Affiliation(s)
- Connor J Cunningham
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roy B Choi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Cardiac-Specific Expression of Cre Recombinase Leads to Age-Related Cardiac Dysfunction Associated with Tumor-like Growth of Atrial Cardiomyocyte and Ventricular Fibrosis and Ferroptosis. Int J Mol Sci 2023; 24:ijms24043094. [PMID: 36834504 PMCID: PMC9962429 DOI: 10.3390/ijms24043094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 02/09/2023] Open
Abstract
Transgenic expression of Cre recombinase driven by a specific promoter is normally used to conditionally knockout a gene in a tissue- or cell-type-specific manner. In αMHC-Cre transgenic mouse model, expression of Cre recombinase is controlled by the myocardial-specific α-myosin heavy chain (αMHC) promoter, which is commonly used to edit myocardial-specific genes. Toxic effects of Cre expression have been reported, including intro-chromosome rearrangements, micronuclei formation and other forms of DNA damage, and cardiomyopathy was observed in cardiac-specific Cre transgenic mice. However, mechanisms associated with Cardiotoxicity of Cre remain poorly understood. In our study, our data unveiled that αMHC-Cre mice developed arrhythmias and died after six months progressively, and none of them survived more than one year. Histopathological examination showed that αMHC-Cre mice had aberrant proliferation of tumor-like tissue in the atrial chamber extended from and vacuolation of ventricular myocytes. Furthermore, the αMHC-Cre mice developed severe cardiac interstitial and perivascular fibrosis, accompanied by significant increase of expression levels of MMP-2 and MMP-9 in the cardiac atrium and ventricular. Moreover, cardiac-specific expression of Cre led to disintegration of the intercalated disc, along with altered proteins expression of the disc and calcium-handling abnormality. Comprehensively, we identified that the ferroptosis signaling pathway is involved in heart failure caused by cardiac-specific expression of Cre, on which oxidative stress results in cytoplasmic vacuole accumulation of lipid peroxidation on the myocardial cell membrane. Taken together, these results revealed that cardiac-specific expression of Cre recombinase can lead to atrial mesenchymal tumor-like growth in the mice, which causes cardiac dysfunction, including cardiac fibrosis, reduction of the intercalated disc and cardiomyocytes ferroptosis at the age older than six months in mice. Our study suggests that αMHC-Cre mouse models are effective in young mice, but not in old mice. Researchers need to be particularly careful when using αMHC-Cre mouse model to interpret those phenotypic impacts of gene responses. As the Cre-associated cardiac pathology matched mostly to that of the patients, the model could also be employed for investigating age-related cardiac dysfunction.
Collapse
|
6
|
Rashbrook VS, Brash JT, Ruhrberg C. Cre toxicity in mouse models of cardiovascular physiology and disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:806-816. [PMID: 37692772 PMCID: PMC7615056 DOI: 10.1038/s44161-022-00125-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 09/12/2023]
Abstract
The Cre-LoxP system provides a widely used method for studying gene requirements in the mouse as the main mammalian genetic model organism. To define the molecular and cellular mechanisms that underlie cardiovascular development, function and disease, various mouse strains have been engineered that allow Cre-LoxP-mediated gene targeting within specific cell types of the cardiovascular system. Despite the usefulness of this system, evidence is accumulating that Cre activity can have toxic effects in cells, independently of its ability to recombine pairs of engineered LoxP sites in target genes. Here, we have gathered published evidence for Cre toxicity in cells and tissues relevant to cardiovascular biology and provide an overview of mechanisms proposed to underlie Cre toxicity. Based on this knowledge, we propose that each study utilising the Cre-LoxP system to investigate gene function in the cardiovascular system should incorporate appropriate controls to account for Cre toxicity.
Collapse
Affiliation(s)
- Victoria S. Rashbrook
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T. Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
7
|
Erben L, Welday JP, Murphy R, Buonanno A. Toxic and Phenotypic Effects of AAV_Cre Used to Transduce Mesencephalic Dopaminergic Neurons. Int J Mol Sci 2022; 23:9462. [PMID: 36012727 PMCID: PMC9408874 DOI: 10.3390/ijms23169462] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
A popular approach to spatiotemporally target genes using the loxP/Cre recombination system is stereotaxic microinjection of adeno-associated virus (AAV) expressing Cre recombinase (AAV_Cre) in specific neuronal structures. Here, we report that AAV_Cre microinjection in the ventral tegmental area (VTA) of ErbB4 Cyt-1-floxed (ErbB4 Cyt-1fl/fl) mice at titers commonly used in the literature (~1012-1013 GC/mL) can have neurotoxic effects on dopaminergic neurons and elicit behavioral abnormalities. However, these effects of AAV_Cre microinjection are independent of ErbB4 Cyt-1 recombination because they are also observed in microinjected wild-type (WT) controls. Mice microinjected with AAV_Cre (1012-1013 GC/mL) exhibit reductions of tyrosine hydroxylase (TH) and dopamine transporter (DAT) expression, loss of dopaminergic neurons, and they behaviorally become hyperactive, fail to habituate in the open field and exhibit sensorimotor gating deficits compared to controls microinjected with AAV_GFP. Importantly, these AAV_Cre non-specific effects are: (1) independent of serotype, (2) occur with vectors expressing either Cre or Cre-GFP fusion protein and (3) preventable by reducing viral titers by 1000-fold (1010 GC/mL), which retains sufficient recombination activity to target floxed genes. Our studies emphasize the importance of including AAV_Cre-injected WT controls in experiments because recombination-independent effects on gene expression, neurotoxicity and behaviors could be erroneously attributed to consequences of gene ablation.
Collapse
Affiliation(s)
| | | | | | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Adkins-Threats M, Mills JC. Cell plasticity in regeneration in the stomach and beyond. Curr Opin Genet Dev 2022; 75:101948. [PMID: 35809361 PMCID: PMC10378711 DOI: 10.1016/j.gde.2022.101948] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Recent studies using cell lineage-tracing techniques, organoids, and single-cell RNA sequencing analyses have revealed: 1) adult organs use cell plasticity programs to recruit progenitor cells to regenerate tissues after injury, and 2) plasticity is far more common than previously thought, even in homeostasis. Here, we focus on the complex interplay of normal stem cell differentiation and plasticity in homeostasis and after injury, using the gastric epithelium as a touchstone. We also examine common features of regenerative programs and discuss the evolutionarily conserved, stepwise process of paligenosis which reprograms mature cells into progenitors that can repair damaged tissue. Finally, we discuss how conserved plasticity programs may help us better understand pathological processes like metaplasia.
Collapse
Affiliation(s)
- Mahliyah Adkins-Threats
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, USA. https://twitter.com/@madkinsthreats
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, USA; Department of Pathology & Immunology, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA.
| |
Collapse
|
9
|
Expression of Cre recombinase in chondrocytes causes abnormal craniofacial and skeletal development. Transgenic Res 2022; 31:399-411. [DOI: 10.1007/s11248-022-00308-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 04/07/2022] [Indexed: 10/18/2022]
|
10
|
Das S, Feng Q, Balasubramanian I, Lin X, Liu H, Pellón-Cardenas O, Yu S, Zhang X, Liu Y, Wei Z, Bonder EM, Verzi MP, Hsu W, Zhang L, Wang TC, Gao N. Colonic healing requires Wnt produced by epithelium as well as Tagln+ and Acta2+ stromal cells. Development 2022; 149:273689. [PMID: 34910127 PMCID: PMC8881740 DOI: 10.1242/dev.199587] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/24/2021] [Indexed: 01/14/2023]
Abstract
Although Wnt signaling is clearly important for the intestinal epithelial homeostasis, the relevance of various sources of Wnt ligands themselves remains incompletely understood. Blocking the release of Wnt in distinct stromal cell types suggests obligatory functions of several stromal cell sources and yields different observations. The physiological contribution of epithelial Wnt to tissue homeostasis remains unclear. We show here that blocking epithelial Wnts affects colonic Reg4+ epithelial cell differentiation and impairs colonic epithelial regeneration after injury in mice. Single-cell RNA analysis of intestinal stroma showed that the majority of Wnt-producing cells were contained in transgelin (Tagln+) and smooth muscle actin α2 (Acta2+) expressing populations. We genetically attenuated Wnt production from these stromal cells using Tagln-Cre and Acta2-CreER drivers, and found that blockage of Wnt release from either epithelium or Tagln+ and Acta2+ stromal cells impaired colonic epithelial healing after chemical-induced injury. Aggregated blockage of Wnt release from both epithelium and Tagln+ or Acta2+ stromal cells drastically diminished epithelial repair, increasing morbidity and mortality. These results from two uncharacterized stromal populations suggested that colonic recovery from colitis-like injury depends on multiple Wnt-producing sources.
Collapse
Affiliation(s)
- Soumyashree Das
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | | | - Xiang Lin
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Haoran Liu
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | | | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Yue Liu
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Edward M. Bonder
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Michael P. Verzi
- Department of Genetics, Rutgers University, Piscataway, New Jersey, USA,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Wei Hsu
- Department of Biomedical Genetics, Center for Oral Biology, James P Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 04642, USA
| | - Lanjing Zhang
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA,Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08536, USA
| | - Timothy C. Wang
- Department of Medicine, Division of Digestive and Liver Diseases, Irving Cancer Research Center, Columbia University, New York, NY 10027, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA,Author for correspondence ()
| |
Collapse
|
11
|
Killy B, Bodendorfer B, Mages J, Ritter K, Schreiber J, Hölscher C, Pracht K, Ekici A, Jäck HM, Lang R. DGCR8 deficiency impairs macrophage growth and unleashes the interferon response to mycobacteria. Life Sci Alliance 2021; 4:4/6/e202000810. [PMID: 33771876 PMCID: PMC8008949 DOI: 10.26508/lsa.202000810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
The mycobacterial cell wall glycolipid trehalose-6,6-dimycolate (TDM) activates macrophages through the C-type lectin receptor MINCLE. Regulation of innate immune cells relies on miRNAs, which may be exploited by mycobacteria to survive and replicate in macrophages. Here, we have used macrophages deficient in the microprocessor component DGCR8 to investigate the impact of miRNA on the response to TDM. Deletion of DGCR8 in bone marrow progenitors reduced macrophage yield, but did not block macrophage differentiation. DGCR8-deficient macrophages showed reduced constitutive and TDM-inducible miRNA expression. RNAseq analysis revealed that they accumulated primary miRNA transcripts and displayed a modest type I IFN signature at baseline. Stimulation with TDM in the absence of DGCR8 induced overshooting expression of IFNβ and IFN-induced genes, which was blocked by antibodies to type I IFN. In contrast, signaling and transcriptional responses to recombinant IFNβ were unaltered. Infection with live Mycobacterium bovis Bacille Calmette-Guerin replicated the enhanced IFN response. Together, our results reveal an essential role for DGCR8 in curbing IFNβ expression macrophage reprogramming by mycobacteria.
Collapse
Affiliation(s)
- Barbara Killy
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Bodendorfer
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kristina Ritter
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany
| | - Jonathan Schreiber
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Hölscher
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany.,German Center for Infection Research (DZIF), Partner Site Borstel, Borstel, Germany
| | - Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Miao ZF, Sun JX, Adkins-Threats M, Pang MJ, Zhao JH, Wang X, Tang KW, Wang ZN, Mills JC. DDIT4 Licenses Only Healthy Cells to Proliferate During Injury-induced Metaplasia. Gastroenterology 2021; 160:260-271.e10. [PMID: 32956680 PMCID: PMC7857017 DOI: 10.1053/j.gastro.2020.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS In stomach, metaplasia can arise from differentiated chief cells that become mitotic via paligenosis, a stepwise program. In paligenosis, mitosis initiation requires reactivation of the cellular energy hub mTORC1 after initial mTORC1 suppression by DNA damage induced transcript 4 (DDIT4 aka REDD1). Here, we use DDIT4-deficient mice and human cells to study how metaplasia increases tumorigenesis risk. METHODS A tissue microarray of human gastric tissue specimens was analyzed by immunohistochemistry for DDIT4. C57BL/6 mice were administered combinations of intraperitoneal injections of high-dose tamoxifen (TAM) to induce spasmolytic polypeptide-expressing metaplasia (SPEM) and rapamycin to block mTORC1 activity, and N-methyl-N-nitrosourea (MNU) in drinking water to induce spontaneous gastric tumors. Stomachs were analyzed for proliferation, DNA damage, and tumor formation. CRISPR/Cas9-generated DDIT4-/- and control human gastric cells were analyzed for growth in vitro and in xenografts with and without 5-fluorouracil (5-FU) treatment. RESULTS DDIT4 was expressed in normal gastric chief cells in mice and humans and decreased as chief cells became metaplastic. Paligenotic Ddit4-/- chief cells maintained constitutively high mTORC1, causing increased mitosis of metaplastic cells despite DNA damage. Lower DDIT4 expression correlated with longer survival of patients with gastric cancer. 5-FU-treated DDIT4-/- human gastric epithelial cells had significantly increased cells entering mitosis despite DNA damage and increased proliferation in vitro and in xenografts. MNU-treated Ddit4-/- mice had increased spontaneous tumorigenesis after multiple rounds of paligenosis induced by TAM. CONCLUSIONS During injury-induced metaplastic proliferation, failure of licensing mTORC1 reactivation correlates with increased proliferation of cells harboring DNA damage, as well as increased tumor formation and growth in mice and humans.
Collapse
Affiliation(s)
- Zhi-Feng Miao
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jing-Xu Sun
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mahliyah Adkins-Threats
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Min-Jiao Pang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jun-Hua Zhao
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xin Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Kai-Wen Tang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Jason C Mills
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri.
| |
Collapse
|
13
|
Sahai N, Bard AM, Devinsky O, Kalume F. Disordered autonomic function during exposure to moderate heat or exercise in a mouse model of Dravet syndrome. Neurobiol Dis 2020; 147:105154. [PMID: 33144172 DOI: 10.1016/j.nbd.2020.105154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To examine autonomic regulation of core body temperature, heart rate (HR), and breathing rate (BR) in response to moderately elevated ambient temperature or moderate physical exercise in a mouse model of Dravet syndrome (DS). METHODS We studied video-EEG, ECG, respiration, and temperature in mice with global heterozygous Scn1a knockout (KO) (DS mice), interneuron specific Scn1a KO, and wildtype (WT) mice during exposure to increased environmental temperature and moderate treadmill exercise. RESULTS Core body temperatures of WT and DS mice were similar during baseline. After 15 mins of heat exposure, the peak value was lower in DS than WT mice. In the following mins of heat exposure, the temperature slowly returned close to baseline level in WT, whereas it remained elevated in DS mice. KO of Scn1a in GABAergic neurons caused similar thermoregulatory deficits in mice. During exercise, the HR increase was less prominent in DS than WT mice. After exercise, the HR was significantly more suppressed in DS. The heart rate variability (HRV) was lower in DS than WT mice during baseline and higher in DS during exercise-recovery periods. SIGNIFICANCE We found novel abnormalities that expand the spectrum of interictal, ictal, and postictal autonomic dysregulation in DS mice. During mild heat stress, there was a significantly blunted correction of body temperature, and a less suppression of both HR and respiration rate in DS than WT mice. These effects were seen in mice with selective KO of Scn1A in GABAergic neurons. During exercise stress, there was diminished increase in HR, followed by an exaggerated HR suppression and HRV elevation during recovery in DS mice compared to controls. These findings suggest that different environmental stressors can uncover distinct autonomic disturbances in DS mice. Interneurons play an important role in thermoregulation. Understanding the spectrum and mechanisms of autonomic disorders in DS may help develop more effective strategies to prevent seizures and SUDEP.
Collapse
Affiliation(s)
- Nikhil Sahai
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Angela M Bard
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Franck Kalume
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
14
|
McKinley KL, Castillo-Azofeifa D, Klein OD. Tools and Concepts for Interrogating and Defining Cellular Identity. Cell Stem Cell 2020; 26:632-656. [PMID: 32386555 PMCID: PMC7250495 DOI: 10.1016/j.stem.2020.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Defining the mechanisms that generate specialized cell types and coordinate their functions is critical for understanding organ development and renewal. New tools and discoveries are challenging and refining our definitions of a cell type. A rapidly growing toolkit for single-cell analyses has expanded the number of markers that can be assigned to a cell simultaneously, revealing heterogeneity within cell types that were previously regarded as homogeneous populations. Additionally, cell types defined by specific molecular markers can exhibit distinct, context-dependent functions; for example, between tissues in homeostasis and those responding to damage. Here we review the current technologies used to identify and characterize cells, and we discuss how experimental and pathological perturbations are adding increasing complexity to our definitions of cell identity.
Collapse
Affiliation(s)
- Kara L McKinley
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - David Castillo-Azofeifa
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Mobley CB, Vechetti IJ, Valentino TR, McCarthy JJ. CORP: Using transgenic mice to study skeletal muscle physiology. J Appl Physiol (1985) 2020; 128:1227-1239. [PMID: 32105520 DOI: 10.1152/japplphysiol.00021.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The development of tissue-specific inducible transgenic mice has provided a powerful tool to study gene function and cell biology in almost any tissue of interest at any given time within the animal's life. The purpose of this review is to describe how to use two different inducible transgenic systems, the Cre-loxP system and the Tet-ON/OFF system, that can be used to study skeletal muscle physiology. Myofiber- and satellite cell-specific Cre-loxP transgenic mice are described as is how these mice can be used to knockout a gene of interest or to deplete satellite cells in adult skeletal muscle, respectively. A myofiber-specific Tet-ON system is described as is how such mice can be used to overexpress a gene of interest or to label myonuclei. How to effectively breed and genotype the transgenic mice are also described in detail. The hope is this review will provide the basic information necessary to facilitate the incorporation of tissue-specific inducible transgenic mice into a skeletal muscle research program.
Collapse
Affiliation(s)
- C Brooks Mobley
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Ivan J Vechetti
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Taylor R Valentino
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
16
|
Zhang Y, Que J. BMP Signaling in Development, Stem Cells, and Diseases of the Gastrointestinal Tract. Annu Rev Physiol 2020; 82:251-273. [PMID: 31618602 DOI: 10.1146/annurev-physiol-021119-034500] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bone morphogenetic protein (BMP) pathway is essential for the morphogenesis of multiple organs in the digestive system. Abnormal BMP signaling has also been associated with disease initiation and progression in the gastrointestinal (GI) tract and associated organs. Recent studies using animal models, tissue organoids, and human pluripotent stem cells have significantly expanded our understanding of the roles played by BMPs in the development and homeostasis of GI organs. It is clear that BMP signaling regulates GI function and disease progression that involve stem/progenitor cells and inflammation in a tissue-specific manner. In this review we discuss these new findings with a focus on the esophagus, stomach, and intestine.
Collapse
Affiliation(s)
- Yongchun Zhang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA; .,Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA; .,Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
17
|
Burclaff J, Willet SG, Sáenz JB, Mills JC. Proliferation and Differentiation of Gastric Mucous Neck and Chief Cells During Homeostasis and Injury-induced Metaplasia. Gastroenterology 2020; 158:598-609.e5. [PMID: 31589873 PMCID: PMC7010566 DOI: 10.1053/j.gastro.2019.09.037] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Adult zymogen-producing (zymogenic) chief cells (ZCs) in the mammalian gastric gland base are believed to arise from descending mucous neck cells, which arise from stem cells. Gastric injury, such as from Helicobacter pylori infection in patients with chronic atrophic gastritis, can cause metaplasia, characterized by gastric cell expression of markers of wound-healing; these cells are called spasmolytic polypeptide-expressing metaplasia (SPEM) cells. We investigated differentiation and proliferation patterns of neck cells, ZCs, and SPEM cells in mice. METHODS C57BL/6 mice were given intraperitoneal injections of high-dose tamoxifen to induce SPEM or gavaged with H pylori (PMSS1) to induce chronic gastric injury. Mice were then given pulses of 5-bromo-2'-deoxyuridine (BrdU) in their drinking water, followed by chase periods without BrdU, or combined with intraperitoneal injections of 5-ethynyl-2'-deoxyuridine. We collected gastric tissues and performed immunofluorescence and immunohistochemical analyses to study gastric cell proliferation, differentiation, and turnover. RESULTS After 8 weeks of continuous BrdU administration, fewer than 10% of homeostatic ZCs incorporated BrdU, whereas 88% of neck cells were labeled. In pulse-chase experiments, various chase periods decreased neck cell label but did not increase labeling of ZCs. When mice were given BrdU at the same time as tamoxifen, more than 90% of cells were labeled in all gastric lineages. After 3 months' recovery (no tamoxifen), ZCs became the predominant BrdU-labeled population, whereas other cells, including neck cells, were mostly negative. When we tracked the labeled cells in such mice over time, we observed that the proportion of BrdU-positive ZCs remained greater than 60% up to 11 months. In mice whose ZCs were the principal BrdU-positive population, acute injury by tamoxifen or chronic injury by H pylori infection resulted in SPEM cells becoming the principal BrdU-positive population. After withdrawal of tamoxifen, BrdU-positive ZCs reappeared. CONCLUSIONS We studied mice in homeostasis or with tamoxifen- or H pylori-induced SPEM. Our findings indicated that mucous neck cells do not contribute substantially to generation of ZCs during homeostasis and that ZCs maintain their own census, likely through infrequent self-replication. After metaplasia-inducing injury, ZCs can become SPEM cells, and then redifferentiate into ZCs on injury resolution.
Collapse
Affiliation(s)
- Joseph Burclaff
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Spencer G Willet
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - José B Sáenz
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Jason C Mills
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri; Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri.
| |
Collapse
|
18
|
Seidlitz T, Chen YT, Uhlemann H, Schölch S, Kochall S, Merker SR, Klimova A, Hennig A, Schweitzer C, Pape K, Baretton GB, Welsch T, Aust DE, Weitz J, Koo BK, Stange DE. Mouse Models of Human Gastric Cancer Subtypes With Stomach-Specific CreERT2-Mediated Pathway Alterations. Gastroenterology 2019; 157:1599-1614.e2. [PMID: 31585123 PMCID: PMC6902245 DOI: 10.1053/j.gastro.2019.09.026] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Patterns of genetic alterations characterize different molecular subtypes of human gastric cancer. We aimed to establish mouse models of these subtypes. METHODS We searched databases to identify genes with unique expression in the stomach epithelium, resulting in the identification of Anxa10. We generated mice with tamoxifen-inducible Cre recombinase (CreERT2) in the Anxa10 gene locus. We created 3 mouse models with alterations in pathways that characterize the chromosomal instability (CIN) and the genomically stable (GS) subtypes of human gastric cancer: Anxa10-CreERT2;KrasG12D/+;Tp53R172H/+;Smad4fl/f (CIN mice), Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Smad4fl/fl (GS-TGBF mice), and Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Apcfl/fl (GS-Wnt mice). We analyzed tumors that developed in these mice by histology for cell types and metastatic potential. We derived organoids from the tumors and tested their response to chemotherapeutic agents and the epithelial growth factor receptor signaling pathway inhibitor trametinib. RESULTS The gastric tumors from the CIN mice had an invasive phenotype and formed liver and lung metastases. The tumor cells had a glandular morphology, similar to human intestinal-type gastric cancer. The gastric tumors from the GS-TGFB mice were poorly differentiated with diffuse morphology and signet ring cells, resembling human diffuse-type gastric cancer. Cells from these tumors were invasive, and mice developed peritoneal carcinomatosis and lung metastases. GS-Wnt mice developed adenomatous tooth-like gastric cancer. Organoids derived from tumors of GS-TGBF and GS-Wnt mice were more resistant to docetaxel, whereas organoids from the CIN tumors were more resistant to trametinib. CONCLUSIONS Using a stomach-specific CreERT2 system, we created mice that develop tumors with morphologic similarities to subtypes of human gastric cancer. These tumors have different patterns of local growth, metastasis, and response to therapeutic agents. They can be used to study different subtypes of human gastric cancer.
Collapse
Affiliation(s)
- Therese Seidlitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Yi-Ting Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Heike Uhlemann
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schölch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Susan Kochall
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sebastian R. Merker
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anna Klimova
- Institute for Medical Informatics and Biometry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Core Unit for Data Management and Analytics (CDMA), National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Alexander Hennig
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Christine Schweitzer
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristin Pape
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gustavo B. Baretton
- German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,Core Unit for Molecular Tumour Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany,Institute of Pathology and Tumour and Normal Tissue Bank of the University Cancer Center, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Thilo Welsch
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniela E. Aust
- German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,Core Unit for Molecular Tumour Diagnostics, National Center for Tumor Diseases (NCT), Dresden, Germany,Institute of Pathology and Tumour and Normal Tissue Bank of the University Cancer Center, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,National Center for Tumor Diseases, Dresden, Germany
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Daniel E. Stange
- Department of Visceral, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,German Cancer Consortium (DKTK), Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany,National Center for Tumor Diseases, Dresden, Germany,Reprint requests Address requests for reprints to: Daniel E. Strange, MD, PhD, Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
19
|
Frahsek M, Schulte K, Chia-Gil A, Djudjaj S, Schueler H, Leuchtle K, Smeets B, Dijkman H, Floege J, Moeller MJ. Cre recombinase toxicity in podocytes: a novel genetic model for FSGS in adolescent mice. Am J Physiol Renal Physiol 2019; 317:F1375-F1382. [PMID: 31588799 DOI: 10.1152/ajprenal.00573.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Here, we show that inducible overexpression of Cre recombinase in glomerular podocytes but not in parietal epithelial cells may trigger focal segmental glomerulosclerosis (FSGS) in juvenile transgenic homocygous Pod-rtTA/LC1 mice. Administration of doxycycline shortly after birth, but not at any other time point later in life, resulted in podocyte injury and development of classical FSGS lesions in these mice. Sclerotic lesions were formed as soon as 3 wk of age, and FSGS progressed with low variability until 13 wk of age. In addition, our experiments identified Cre toxicity as a potentially relevant limitation for studies in podocytes of transgenic animals. In summary, our study establishes a novel genetic model for FSGS in mice, which exhibits low variability and manifests already at a young age.
Collapse
Affiliation(s)
- Madeleine Frahsek
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Kevin Schulte
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany.,Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Arnaldo Chia-Gil
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Herdit Schueler
- Institute of Human Genetics, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Katja Leuchtle
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University, Nijmegen, The Netherlands
| | - Henry Dijkman
- Department of Pathology, Radboud University, Nijmegen, The Netherlands
| | - Jürgen Floege
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Marcus J Moeller
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany.,Heisenberg Chair for Preventive and Translational Nephrology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
20
|
Drake KA, Fessler AR, Carroll TJ. Methods for renal lineage tracing: In vivo and beyond. Methods Cell Biol 2019; 154:121-143. [PMID: 31493814 DOI: 10.1016/bs.mcb.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lineage tracing has resulted in fundamental discoveries in kidney development and disease and remains a powerful technique to study mechanisms of organogenesis, homeostasis, and repair/regeneration. Following decades of research on the cellular and molecular regulation of renal organogenesis, the kidney has become one of the most well-characterized organs, resulting in exciting advancements in pluripotent stem cell differentiation, tissue bioengineering, and the potential for developing novel regenerative therapies for kidney disease. Lineage tracing, or the labeling of progeny cells arising from a single cell or group of cells, allows for spatial and temporal analyses of dynamic in vivo and in vitro processes. As lineage tracing techniques expand across disciplines of developmental biology, stem cell biology, and regenerative medicine, careful experimental design and interpretation, along with an understanding of the basic principles and technical limitations, are essential for utilizing genetically complex lineage tracing models to further understand kidney development and disease.
Collapse
Affiliation(s)
- Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alicia R Fessler
- Department of Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Thomas J Carroll
- Department of Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
21
|
Eckrich S, Hecker D, Sorg K, Blum K, Fischer K, Münkner S, Wenzel G, Schick B, Engel J. Cochlea-Specific Deletion of Ca v1.3 Calcium Channels Arrests Inner Hair Cell Differentiation and Unravels Pitfalls of Conditional Mouse Models. Front Cell Neurosci 2019; 13:225. [PMID: 31178698 PMCID: PMC6538774 DOI: 10.3389/fncel.2019.00225] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022] Open
Abstract
Inner hair cell (IHC) Cav1.3 Ca2+ channels are multifunctional channels mediating Ca2+ influx for exocytosis at ribbon synapses, the generation of Ca2+ action potentials in pre-hearing IHCs and gene expression. IHCs of deaf systemic Cav1.3-deficient (Cav1.3-/-) mice stay immature because they fail to up-regulate voltage- and Ca2+-activated K+ (BK) channels but persistently express small conductance Ca2+-activated K+ (SK2) channels. In pre-hearing wildtype mice, cholinergic neurons from the superior olivary complex (SOC) exert efferent inhibition onto spontaneously active immature IHCs by activating their SK2 channels. Because Cav1.3 plays an important role for survival, health and function of SOC neurons, SK2 channel persistence and lack of BK channels in systemic Cav1.3-/- IHCs may result from malfunctioning neurons of the SOC. Here we analyze cochlea-specific Cav1.3 knockout mice with green fluorescent protein (GFP) switch reporter function, Pax2::cre;Cacna1d-eGFPflex/flexand Pax2::cre;Cacna1d-eGFPflex/-. Profound hearing loss, lack of BK channels and persistence of SK2 channels in Pax2::cre;Cacna1d-eGFPflex/- mice recapitulated the phenotype of systemic Cav1.3-/- mice, indicating that in wildtype mice, regulation of SK2 and BK channel expression is independent of Cav1.3 expression in SOC neurons. In addition, we noticed dose-dependent GFP toxicity leading to death of basal coil IHCs of Pax2::cre;Cacna1d-eGFPflex/flex mice, likely because of high GFP concentration and small repair capacity. This and the slower time course of Pax2-driven Cre recombinase in switching two rather than one Cacna1d-eGFPflex allele lead us to study Pax2::cre;Cacna1d-eGFPflex/- mice. Notably, control Cacna1d-eGFPflex/- IHCs showed a significant reduction in Cav1.3 channel cluster sizes and currents, suggesting that the intronic construct interfered with gene translation or splicing. These pitfalls are likely to be a frequent problem of many genetically modified mice with complex or multiple gene-targeting constructs or fluorescent proteins. Great caution and appropriate controls are therefore required.
Collapse
Affiliation(s)
- Stephanie Eckrich
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Dietmar Hecker
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Katharina Sorg
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Kerstin Fischer
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Stefan Münkner
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| | - Gentiana Wenzel
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Bernhard Schick
- Department of Otorhinolaryngology, Saarland University, Homburg, Germany
| | - Jutta Engel
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
22
|
Rezai Amin S, Gruszczynski C, Guiard BP, Callebert J, Launay JM, Louis F, Betancur C, Vialou V, Gautron S. Viral vector-mediated Cre recombinase expression in substantia nigra induces lesions of the nigrostriatal pathway associated with perturbations of dopamine-related behaviors and hallmarks of programmed cell death. J Neurochem 2019; 150:330-340. [PMID: 30748001 DOI: 10.1111/jnc.14684] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 02/03/2023]
Abstract
Cre/loxP recombination is a widely used approach to study gene function in vivo, using mice models expressing the Cre recombinase under the control of specific promoters or through viral delivery of Cre-expressing constructs. A profuse literature on transgenic mouse lines points out the deleterious effects of Cre expression in various cell types and tissues, presumably by acting on illegitimate loxP-like sites present in the genome. However, most studies reporting the consequences of Cre-lox gene invalidation often omit adequate controls to exclude the potential toxic effects of Cre, compromising the interpretation of data. In this study, we report the anatomical, neurochemical, and behavioral consequences in mice of adeno-associated virus (AAV)-mediated Cre expression in the dopaminergic nuclei substantia nigra, at commonly used viral titers (3 × 109 genome copies/0.3 μL or 2 × 109 genome copies/0.6 μL). We found that injecting AAV-eGFP-Cre into the SN engendered drastic and reproducible modifications of behavior, with increased basal locomotor activity as well as impaired locomotor response to cocaine compared to AAV-eGFP-injected controls. Cre expression in the SN induced a massive decrease in neuronal populations of both pars compacta and pars reticulata and dopamine depletion in the nigrostriatal pathway. This anatomical injury was associated with typical features of programmed cell death, including an increase in DNA break markers, evidence of apoptosis, and disrupted macroautophagy. These observations underscore the need for careful control of Cre toxicity in the brain and the reassessment of previous studies. In addition, our findings suggest that Cre-mediated ablation may constitute an efficient tool to explore the function of specific cell populations and areas in the brain, and the impact of neurodegeneration in these populations.
Collapse
Affiliation(s)
- Sara Rezai Amin
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Carole Gruszczynski
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Bruno P Guiard
- Université de Toulouse, CNRS, Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Toulouse, France
| | - Jacques Callebert
- INSERM U942, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Jean-Marie Launay
- INSERM U942, Hôpital Lariboisière, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Franck Louis
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Vincent Vialou
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Sophie Gautron
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
23
|
Balkawade RS, Chen C, Crowley MR, Crossman DK, Clapp WL, Verlander JW, Marshall CB. Podocyte-specific expression of Cre recombinase promotes glomerular basement membrane thickening. Am J Physiol Renal Physiol 2019; 316:F1026-F1040. [PMID: 30810063 DOI: 10.1152/ajprenal.00359.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Conditional gene targeting using Cre recombinase has offered a powerful tool to modify gene function precisely in defined cells/tissues and at specific times. However, in mammalian cells, Cre recombinase can be genotoxic. The importance of including Cre-expressing control mice to avoid misinterpretation and to maximize the validity of the experimental results has been increasingly recognized. While studying the role of podocytes in the pathogenesis of glomerular basement membrane (GBM) thickening, we used Cre recombinase driven by the podocyte-specific podocin promoter (NPHS2-Cre) to generate a conditional knockout. By conventional structural and functional measures (histology by periodic acid-Schiff staining, albuminuria, and plasma creatinine), we did not detect significant differences between NPHS2-Cre transgenic and wild-type control mice. However, surprisingly, the group that expressed Cre transgene alone developed signs of podocyte toxicity, including marked GBM thickening, loss of normal foot process morphology, and reduced Wilms tumor 1 expression. GBM thickening was characterized by altered expression of core structural protein laminin isoform α5β2γ1. RNA sequencing analysis of extracted glomeruli identified 230 genes that were significant and differentially expressed (applying a q < 0.05-fold change ≥ ±2 cutoff) in NPHS2-Cre mice compared with wild-type control mice. Many biological processes were reflected in the RNA sequencing data, including regulation of the extracellular matrix and pathways related to apoptosis and cell death. This study highlights the importance of including the appropriate controls for potential Cre-mediated toxicity in conditional gene-targeting experiments. Indeed, omitting the Cre transgene control can result in critical errors during interpretation of experimental data.
Collapse
Affiliation(s)
- Rohan S Balkawade
- Department of Veterans Affairs Medical Center , Birmingham, Alabama.,Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Chao Chen
- Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine Electron Microscopy Core, University of Florida , Gainesville, Florida
| | - Michael R Crowley
- Heflin Center for Genomic Science, Department of Genetics, University of Alabama at Birmingham , Birmingham, Alabama
| | - David K Crossman
- Heflin Center for Genomic Science, Department of Genetics, University of Alabama at Birmingham , Birmingham, Alabama
| | - William L Clapp
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida , Gainesville, Florida
| | - Jill W Verlander
- Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine Electron Microscopy Core, University of Florida , Gainesville, Florida
| | - Caroline B Marshall
- Department of Veterans Affairs Medical Center , Birmingham, Alabama.,Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
24
|
Bohin N, Carlson EA, Samuelson LC. Genome Toxicity and Impaired Stem Cell Function after Conditional Activation of CreER T2 in the Intestine. Stem Cell Reports 2018; 11:1337-1346. [PMID: 30449703 PMCID: PMC6294112 DOI: 10.1016/j.stemcr.2018.10.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
With the tamoxifen-inducible CreERT2 system, genetic recombination can be temporally controlled in a cell-type-specific manner in intact animals, permitting dissection of the molecular underpinnings of mammalian physiology. Here we present a significant drawback to CreERT2 technology for analysis of intestinal stem cells. Using the intestine-specific Villin-CreERT2 mouse strain, we observed delayed intestinal regeneration post irradiation. Villin-CreERT2 activation was associated with DNA damage and cryptic loxP site cleavage. Analysis of stem cell-specific CreERT2 strains showed that the genome toxicity impairs function of crypt base columnar stem cells, resulting in loss of organoid initiating activity. Importantly, the stem cell impairment is short-lived, with return to normal by 7 days post tamoxifen treatment. Our findings demonstrate that mouse genetic experiments that utilize CreERT2 should consider the confounding effects of enhanced stem cell sensitivity to genome toxicity resulting from CreERT2 activation. Intestinal stem cell (ISC) toxicity induced in mice by CreERT2 activation Impaired organoid formation after activation of ISC-specific CreERT2 strains Genotoxicity and impaired crypt regeneration in Villin-CreERT2 mice Impaired ISC function and genotoxicity repaired by 7 days after activation
Collapse
Affiliation(s)
- Natacha Bohin
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular & Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth A Carlson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Linda C Samuelson
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Cellular & Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
25
|
Abdel Aziz N, Nono JK, Mpotje T, Brombacher F. The Foxp3+ regulatory T-cell population requires IL-4Rα signaling to control inflammation during helminth infections. PLoS Biol 2018; 16:e2005850. [PMID: 30379806 PMCID: PMC6231676 DOI: 10.1371/journal.pbio.2005850] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 11/12/2018] [Accepted: 10/17/2018] [Indexed: 12/27/2022] Open
Abstract
Forkhead box P3 (Foxp3+) regulatory T (Treg)-cell function is controlled by environmental cues of which cytokine-mediated signaling is a dominant component. In vivo, interleukin-4 (IL-4)-mediated signaling via IL-4 receptor alpha (IL-4Rα) mediates Treg cell transdifferentiation into ex-Foxp3 T helper 2 (Th2) or T helper 17 (Th17) cells. However, IL-4-mediated signaling also reinforces the Foxp3 Treg compartment in vitro. We generated Foxp3-specific IL-4Rα-deficient mice and demonstrated differential efficiency of IL-4Rα deletion in male (approximately 90%) and female (approximately 40%) animals, because of cyclic recombinase (Cre)-mediated X-linked foxp3 inactivation. Irrespective of the degree of IL-4Rα deletion within the Foxp3+ Treg cell population, mice showed exacerbation of immune effector responses with aggravated tissue pathology in tissue-dwelling helminth infections (Schistosoma mansoni or Nippostrongylus brasiliensis). Mechanistically, IL-4Rα deletion in males and females led to a reduced expression of Foxp3 and subsequently an impaired accumulation of Foxp3+ Treg cells to inflamed tissues. In-depth cellular typing by flow cytometry revealed that the impairment of IL-4Rα-mediated signaling during helminth infections decreased the ability of central Treg cells to convert into effector Treg (eTreg) cells and caused a significant down-regulation of markers associated with Treg cell migration (C-X-C motif chemokine receptor 3 [CXCR3]) and accumulation in inflamed tissues (GATA binding protein 3 [GATA3]) as well as survival (B cell lymphoma 2 [Bcl-2]). These findings unprecedentedly, to our knowledge, uncover a role for IL-4Rα signaling in the positive regulation of Foxp3+ Treg cell function in vivo. Complementing our past knowledge on a widely reported role for IL-4Rα signaling in the negative regulation and transdifferentiation of Foxp3+ Treg cells in vivo, our present findings reveal the host requirement for an intact, but not reduced or potentiated, IL-4Rα-mediated signaling on Foxp3+ Treg cells to optimally control inflammation during helminth infections. Host soluble mediators such as cytokines play a key role in the regulation of the immune response. Forkhead box P3 (Foxp3+) regulatory T (Treg) cells, which are involved in maintaining self-tolerance and immune system homeostasis, are influenced by cytokines, including interleukin-4 (IL-4). However, opposing reports have emerged on the effect of this cytokine on Treg cells. Some evidence suggests IL-4 inhibits Treg cells, whereas other studies indicate a supportive role for this cytokine in Treg cell biology and function. To unambiguously address this question, we generated mice with IL-4 receptor specifically removed from the Treg cell population. Our newly generated mice did not show any sign of spontaneous inflammation during homeostasis, but when challenged with an experimental infection by parasitic worms, deletion of the IL-4 receptor from the Treg cell population led to increased inflammation and aggravated tissue pathology. Several defects such as poor activation, reduced promigratory marker expression, and reduced survival were apparent in Treg cells with impaired IL-4 responsiveness. Our evidence presents a strong case for a supportive role of IL-4 via IL-4 receptor in the biology and optimal regulatory function of Treg cells during worm infections.
Collapse
Affiliation(s)
- Nada Abdel Aziz
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Biotechnology/Biomolecular Chemistry Program, Chemistry Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Justin Komguep Nono
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- The Medical Research Centre, Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
- * E-mail: (FB); (JKN)
| | - Thabo Mpotje
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa
- University of Cape Town, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- * E-mail: (FB); (JKN)
| |
Collapse
|
26
|
Lrig1 marks a population of gastric epithelial cells capable of long-term tissue maintenance and growth in vitro. Sci Rep 2018; 8:15255. [PMID: 30323305 PMCID: PMC6189208 DOI: 10.1038/s41598-018-33578-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 10/02/2018] [Indexed: 12/16/2022] Open
Abstract
The processes involved in renewal of the epithelium that lines the mouse stomach remain unclear. Apart from the cells in the isthmus, several other populations located deeper in the gastric glands have been suggested to contribute to the maintenance of the gastric epithelium. Here, we reveal that Lrig1 is expressed in the basal layer of the forestomach and the lower part of glands in the corpus and pylorus. In the glandular epithelium of the stomach, Lrig1 marks a heterogeneous population comprising mainly non-proliferative cells. Yet, fate-mapping experiments using a knock-in mouse line expressing Cre specifically in Lrig1+ cells demonstrate that these cells are able to contribute to the long-term maintenance of the gastric epithelium. Moreover, when cultured in vitro, cells expressing high level of Lrig1 have much higher organoid forming potential than the corresponding cellular populations expressing lower levels of Lrig1. Taken together, these observations show that Lrig1 is expressed primarily by differentiated cells, but that these cells can be recruited to contribute to the maintenance of the gastric epithelium. This confirms previous observations that cells located in the lower segments of gastric glands can participate in tissue replenishment.
Collapse
|
27
|
Abstract
Chronic injury and inflammation in the esophagus can cause a change in cellular differentiation known as metaplasia. Most commonly, the differentiation changes manifest as Barrett's esophagus (BE), characterized by the normal stratified squamous epithelium converting into a cuboidal-columnar, glandular morphology. BE cells can phenotypically resemble specific normal cell types of the stomach or intestine, or they can have overlapping phenotypes in disorganized admixtures. The stomach can also undergo metaplasia characterized by aberrant gastric or intestinal differentiation patterns. In both organs, it has been argued that metaplasia may represent a recapitulation of the embryonic or juvenile gastrointestinal tract, as cells access a developmental progenitor genetic program that can help repair damaged tissue. Here, we review the normal development of esophagus and stomach, and describe how BE represents an intermixing of cells resembling gastric pseudopyloric (SPEM) and intestinal metaplasia. We discuss a cellular process recently termed "paligenosis" that governs how mature, differentiated cells can revert to a proliferating progenitor state in metaplasia. We discuss the "Cyclical Hit" theory in which paligenosis might be involved in the increased risk of metaplasia for progression to cancer. However, somatic mutations might occur in proliferative phases and then be warehoused upon redifferentiation. Through years of chronic injury and many rounds of paligenosis and dedifferentiation, eventually a cell with a mutation that prevents dedifferentiation may arise and clonally expand fueling stable metaplasia and potentially thereafter acquiring additional mutations and progressing to dysplasia and cancer.
Collapse
Affiliation(s)
- Ramon U Jin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason C Mills
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
28
|
Wu Q, Han Y, Tong Q. Current Genetic Techniques in Neural Circuit Control of Feeding and Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:211-233. [PMID: 30390293 DOI: 10.1007/978-981-13-1286-1_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current epidemic of obesity and its associated metabolic syndromes imposes unprecedented challenges to our society. Despite intensive research focus on obesity pathogenesis, an effective therapeutic strategy to treat and cure obesity is still lacking. The obesity development is due to a disturbed homeostatic control of feeding and energy expenditure, both of which are controlled by an intricate neural network in the brain. Given the inherent complexity of brain networks in controlling feeding and energy expenditure, the understanding of brain-based pathophysiology for obesity development is limited. One key limiting factor in dissecting neural pathways for feeding and energy expenditure is unavailability of techniques that can be used to effectively reduce the complexity of the brain network to a tractable paradigm, based on which a strong hypothesis can be tested. Excitingly, emerging techniques have been involved to be able to link specific groups of neurons and neural pathways to behaviors (i.e., feeding and energy expenditure). In this chapter, novel techniques especially those based on animal models and viral vector approaches will be discussed. We hope that this chapter will provide readers with a basis that can help to understand the literatures using these techniques and with a guide to apply these exciting techniques to investigate brain mechanisms underlying feeding and energy expenditure.
Collapse
Affiliation(s)
- Qi Wu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Children's Nutrition Research Center, Research Service of Department of Agriculture of USA, Houston, TX, USA.
| | - Yong Han
- Department of Pediatrics, Baylor College of Medicine, USDA-ARS, Houston, TX, USA
| | - Qingchun Tong
- Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
29
|
Saenz JB, Burclaff J, Mills JC. Modeling Murine Gastric Metaplasia Through Tamoxifen-Induced Acute Parietal Cell Loss. Methods Mol Biol 2017; 1422:329-39. [PMID: 27246044 DOI: 10.1007/978-1-4939-3603-8_28] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Parietal cell loss represents the initial step in the sequential progression toward gastric adenocarcinoma. In the setting of chronic inflammation, the expansion of the mucosal response to parietal cell loss characterizes a crucial transition en route to gastric dysplasia. Here, we detail methods for using the selective estrogen receptor modulator tamoxifen as a novel tool to rapidly and reversibly induce parietal cell loss in mice in order to study the mechanisms that underlie these pre-neoplastic events.
Collapse
Affiliation(s)
- Jose B Saenz
- Division of Gastroenterology, Departments of Medicine, Washington University School of Medicine, Box 8124, 660 So. Euclid Ave., St. Louis, MO, 63110, USA
| | - Joseph Burclaff
- Division of Gastroenterology, Departments of Medicine, Washington University School of Medicine, Box 8124, 660 So. Euclid Ave., St. Louis, MO, 63110, USA.,Division of Gastroenterology, Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jason C Mills
- Division of Gastroenterology, Departments of Medicine, Washington University School of Medicine, Box 8124, 660 So. Euclid Ave., St. Louis, MO, 63110, USA. .,Division of Gastroenterology, Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Division of Gastroenterology, Departments of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
30
|
Schwann Cells in Neuromuscular Junction Formation and Maintenance. J Neurosci 2017; 36:9770-81. [PMID: 27656017 DOI: 10.1523/jneurosci.0174-16.2016] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 07/14/2016] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED The neuromuscular junction (NMJ) is a tripartite synapse that is formed by motor nerve terminals, postjunctional muscle membranes, and terminal Schwann cells (TSCs) that cover the nerve-muscle contact. NMJ formation requires intimate communications among the three different components. Unlike nerve-muscle interaction, which has been well characterized, less is known about the role of SCs in NMJ formation and maintenance. We show that SCs in mice lead nerve terminals to prepatterned AChRs. Ablating SCs at E8.5 (i.e., prior nerve arrival at the clusters) had little effect on aneural AChR clusters at E13.5, suggesting that SCs may not be necessary for aneural clusters. SC ablation at E12.5, a time when phrenic nerves approach muscle fibers, resulted in smaller and fewer nerve-induced AChR clusters; however, SC ablation at E15.5 reduced AChR cluster size but had no effect on cluster density, suggesting that SCs are involved in AChR cluster maturation. Miniature endplate potential amplitude, but not frequency, was reduced when SCs were ablated at E15.5, suggesting that postsynaptic alterations may occur ahead of presynaptic deficits. Finally, ablation of SCs at P30, after NMJ maturation, led to NMJ fragmentation and neuromuscular transmission deficits. Miniature endplate potential amplitude was reduced 3 d after SC ablation, but both amplitude and frequency were reduced 6 d after. Together, these results indicate that SCs are not only required for NMJ formation, but also necessary for its maintenance; and postsynaptic function and structure appeared to be more sensitive to SC ablation. SIGNIFICANCE STATEMENT Neuromuscular junctions (NMJs) are critical for survival and daily functioning. Defects in NMJ formation during development or maintenance in adulthood result in debilitating neuromuscular disorders. The role of Schwann cells (SCs) in NMJ formation and maintenance was not well understood. We genetically ablated SCs during development and after NMJ formation to investigate the consequences of the ablation. This study reveals a critical role of SCs in NMJ formation as well as maintenance.
Collapse
|
31
|
Ji H, Lu Y, Shi Y. Seeds in the liver. Acta Histochem 2017; 119:349-356. [PMID: 28389020 DOI: 10.1016/j.acthis.2017.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/28/2017] [Accepted: 03/21/2017] [Indexed: 02/05/2023]
Abstract
The liver is a crucial organ for homeostasis and has a tremendous self-renewal and regenerative capacity. It has long been believed that the self-renewal and repair of the liver within a given physiological condition or its repopulation in chronic liver diseases, when hepatocyte proliferation is impaired, will primarily be conducted by the proliferating duct cells, termed "oval cells" or hepatic progenitor cells (HPCs). In addition, numerous studies have revealed that HPCs are the initial tumor cells of liver cancer under certain micro-environments. However, benefit from the extensive application of lineage tracing strategies using the Cre/LoxP system, researchers have redefined the fate of these bipotential cells, raising obvious controversies regarding the capacity of liver cells to control their own biology and differentiation. Here, we review the relevant articles, focusing on cell-lineage tracing to better understanding seed cells and their distinct fate in the liver.
Collapse
Affiliation(s)
- Hongjie Ji
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, China; Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, China
| | - Yujun Shi
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, China; Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, China.
| |
Collapse
|
32
|
Abstract
The bone morphogenetic proteins, (BMP)s are regulatory peptides that have significant effects on the growth and differentiation of gastrointestinal tissues. In addition, the BMPs have been shown to exert anti-inflammatory actions in the gut and to negatively regulate the growth of gastric neoplasms. The role of BMP signaling in the regulation of gastric metaplasia, dysplasia and neoplasia has been poorly characterized. Transgenic expression in the mouse stomach of the BMP inhibitor noggin leads to decreased parietal cell number, increased epithelial cell proliferation, and to the emergence of SPEM. Moreover, expression of noggin increases Helicobacter-induced inflammation and epithelial cell proliferation, accelerates the development of dysplasia, and it increases the expression of signal transducer and activator of transcription 3 (STAT3) and of activation-induced cytidine deaminase (AID). These findings provide new clues for a better understanding of the pathophysiological mechanisms that regulate gastric inflammation and the development of both dysplastic and neoplastic lesions of the stomach.
Collapse
|
33
|
Lim J, Burclaff J, He G, Mills JC, Long F. Unintended targeting of Dmp1-Cre reveals a critical role for Bmpr1a signaling in the gastrointestinal mesenchyme of adult mice. Bone Res 2017; 5:16049. [PMID: 28163952 PMCID: PMC5282469 DOI: 10.1038/boneres.2016.49] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/10/2016] [Accepted: 09/18/2016] [Indexed: 12/20/2022] Open
Abstract
Cre/loxP technology has been widely used to study cell type-specific functions of genes. Proper interpretation of such data critically depends on a clear understanding of the tissue specificity of Cre expression. The Dmp1-Cre mouse, expressing Cre from a 14-kb DNA fragment of the mouse Dmp1 gene, has become a common tool for studying gene function in osteocytes, but the presumed cell specificity is yet to be fully established. By using the Ai9 reporter line that expresses a red fluorescent protein upon Cre recombination, we find that in 2-month-old mice, Dmp1-Cre targets not only osteocytes within the bone matrix but also osteoblasts on the bone surface and preosteoblasts at the metaphyseal chondro-osseous junction. In the bone marrow, Cre activity is evident in certain stromal cells adjacent to the blood vessels, but not in adipocytes. Outside the skeleton, Dmp1-Cre marks not only the skeletal muscle fibers, certain cells in the cerebellum and the hindbrain but also gastric and intestinal mesenchymal cells that express Pdgfra. Confirming the utility of Dmp1-Cre in the gastrointestinal mesenchyme, deletion of Bmpr1a with Dmp1-Cre causes numerous large polyps along the gastrointestinal tract, consistent with prior work involving inhibition of BMP signaling. Thus, caution needs to be exercised when using Dmp1-Cre because it targets not only the osteoblast lineage at an earlier stage than previously appreciated, but also a number of non-skeletal cell types.
Collapse
Affiliation(s)
- Joohyun Lim
- Department of Orthopaedic Surgery, Washington University School of Medicine , St. Louis, MO, USA
| | - Joseph Burclaff
- Division of Gastroenterology, Departments of Medicine and Pathology and Immunology, Washington University School of Medicine , St. Louis, MO, USA
| | - Guangxu He
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA; Department of Orthopedics, The Second Xiangya Hospital, Central South University, Hunan 410011, China
| | - Jason C Mills
- Division of Gastroenterology, Departments of Medicine and Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Fanxin Long
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
Bartfeld S, Koo BK. Adult gastric stem cells and their niches. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 6. [PMID: 28044412 DOI: 10.1002/wdev.261] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/18/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Adult gastric stem cells replenish the gastric epithelium throughout life. Recent studies have identified diverse populations of stem cells, progenitor cells, and even differentiated cells that can regain stem cell capacity, so highlighting an unexpected plasticity within the gastric epithelium, both in the corpus and antrum. Two niches seem to co-exist in the gastric unit: one in the isthmus region and the other at the base of the gland, although the precise features of the cell populations and the two niches are currently under debate. A variety of gastric organoid models have been established, providing new insights into niche factors required by the gastric stem cell populations. Here we review our current knowledge of gastric stem cell populations, their markers and interactions, important niche factors, and different gastric organoid systems. WIREs Dev Biol 2017, 6:e261. doi: 10.1002/wdev.261 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Sina Bartfeld
- Research Centre for Infectious Diseases, University of Wuerzburg, Wuerzburg, Germany
| | - Bon-Kyoung Koo
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Moore BD, Khurana SS, Huh WJ, Mills JC. Hepatocyte nuclear factor 4α is required for cell differentiation and homeostasis in the adult mouse gastric epithelium. Am J Physiol Gastrointest Liver Physiol 2016; 311:G267-75. [PMID: 27340127 PMCID: PMC5007292 DOI: 10.1152/ajpgi.00195.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 01/31/2023]
Abstract
We have previously shown that the sequential transcription factors Xbp1→Mist1 (Bhlha15) govern the ultrastructural maturation of the secretory apparatus in enzyme-secreting zymogenic chief cells (ZCs) in the gastric unit. Here we sought to identify transcriptional regulators upstream of X-box binding protein 1 (XBP1) and MIST1. We used immunohistochemistry to characterize Hnf4α(flox/flox) adult mouse stomachs after tamoxifen-induced deletion of Hnf4α We used qRT-PCR, Western blotting, and chromatin immunoprecipitation to define the molecular interaction between hepatocyte nuclear factor 4 alpha (HNF4α) and Xbp1 in mouse stomach and human gastric cells. We show that HNF4α protein is expressed in pit (foveolar) cells, mucous neck cells, and zymogenic chief cells (ZCs) of the corpus gastric unit. Loss of HNF4α in adult mouse stomach led to reduced ZC size and ER content, phenocopying previously characterized effects of Xbp1 deletion. However, HNF4α(Δ/Δ) stomachs also exhibited additional phenotypes including increased proliferation in the isthmal stem cell zone and altered mucous neck cell migration, indicating a role of HNF4α in progenitor cells as well as in ZCs. HNF4α directly occupies the Xbp1 promoter locus in mouse stomach, and forced HNF4α expression increased abundance of XBP1 mRNA in human gastric cancer cells. Finally, as expected, loss of HNF4α caused decreased Xbp1 and Mist1 expression in mouse stomachs. We show that HNF4α regulates homeostatic proliferation in the gastric epithelium and is both necessary and sufficient for the upstream regulation of the Xbp1→Mist1 axis in maintenance of ZC secretory architecture.
Collapse
Affiliation(s)
- Benjamin D. Moore
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Shradha S. Khurana
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Won Jae Huh
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| | - Jason C. Mills
- 1Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St. Louis, Missouri
| |
Collapse
|
36
|
Abstract
The stomach, an organ derived from foregut endoderm, secretes acid and enzymes and plays a key role in digestion. During development, mesenchymal-epithelial interactions drive stomach specification, patterning, differentiation and growth through selected signaling pathways and transcription factors. After birth, the gastric epithelium is maintained by the activity of stem cells. Developmental signals are aberrantly activated and stem cell functions are disrupted in gastric cancer and other disorders. Therefore, a better understanding of stomach development and stem cells can inform approaches to treating these conditions. This Review highlights the molecular mechanisms of stomach development and discusses recent findings regarding stomach stem cells and organoid cultures, and their roles in investigating disease mechanisms.
Collapse
Affiliation(s)
- Tae-Hee Kim
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4 Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
37
|
Abstract
Gastric diseases cause considerable worldwide burden. However, the stomach is still poorly understood in terms of the molecular-cellular processes that govern its development and homeostasis. In particular, the complex relationship between the differentiated cell types located within the stomach and the stem and progenitor cells that give rise to them is significantly understudied relative to other organs. In this review, we will highlight the current state of the literature relating to specification of gastric cell lineages from embryogenesis to adulthood. Special emphasis is placed on substantial gaps in knowledge about stomach specification that we think should be tackled to advance the field. For example, it has long been assumed that adult gastric units have a granule-free stem cell that gives rise to all differentiated lineages. Here we will point out that there are also other models that fit all extant data, such as long-lived lineage-committed progenitors that might serve as a source of new cells during homeostasis.
Collapse
Affiliation(s)
- Spencer G. Willet
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jason C. Mills
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
- Correspondence Address correspondence to: Jason C. Mills, MD, PhD, Washington University School of Medicine, Box 8124, 660 South Euclid Avenue, St. Louis, Missouri 63110. fax: (314) 362-7487.Washington University School of MedicineBox 8124, 660 South Euclid AvenueSt. LouisMissouri 63110
| |
Collapse
|
38
|
Mariscal AM, González-González L, Querol E, Piñol J. All-in-one construct for genome engineering using Cre-lox technology. DNA Res 2016; 23:263-70. [PMID: 27084897 PMCID: PMC4909314 DOI: 10.1093/dnares/dsw015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/02/2016] [Indexed: 12/25/2022] Open
Abstract
Mycoplasma genitalium is an appealing model of a minimal cell and synthetic biology study, and it was one of the first organisms whose genome was fully sequenced and chemically synthesized. Despite its usefulness as a model organism, many genetic tools well established for other microorganisms are not currently available in mycoplasmas. We have developed several vectors to adapt the Cre-lox technology for genome engineering in M. genitalium, providing an all-in-one construct that could be also useful to obtain unmarked genetic modifications in many other slow growing microorganisms. This construct contains a modified promoter sequence based in TetR system that exhibits an enhanced control on Cre recombinase expression, virtually abolishing the presence of this recombinase in the absence of inducer. This allows to introduce the Cre recombinase gene and the desired genetic modification in a single transformation step. In addition, this inducible promoter may be a very promising tool for a wide range of molecular applications.
Collapse
Affiliation(s)
- Ana M Mariscal
- Departament de Bioquímica i Biologia Molecular and Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 0819, Spain
| | - Luis González-González
- Departament de Bioquímica i Biologia Molecular and Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 0819, Spain
| | - Enrique Querol
- Departament de Bioquímica i Biologia Molecular and Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 0819, Spain
| | - Jaume Piñol
- Departament de Bioquímica i Biologia Molecular and Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès 0819, Spain
| |
Collapse
|
39
|
New inducible genetic method reveals critical roles of GABA in the control of feeding and metabolism. Proc Natl Acad Sci U S A 2016; 113:3645-50. [PMID: 26976589 DOI: 10.1073/pnas.1602049113] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Currently available inducible Cre/loxP systems, despite their considerable utility in gene manipulation, have pitfalls in certain scenarios, such as unsatisfactory recombination rates and deleterious effects on physiology and behavior. To overcome these limitations, we designed a new, inducible gene-targeting system by introducing an in-frame nonsense mutation into the coding sequence of Cre recombinase (nsCre). Mutant mRNAs transcribed from nsCre transgene can be efficiently translated into full-length, functional Cre recombinase in the presence of nonsense suppressors such as aminoglycosides. In a proof-of-concept model, GABA signaling from hypothalamic neurons expressing agouti-related peptide (AgRP) was genetically inactivated within 4 d after treatment with a synthetic aminoglycoside. Disruption of GABA synthesis in AgRP neurons in young adult mice led to a dramatic loss of body weight due to reduced food intake and elevated energy expenditure; they also manifested glucose intolerance. In contrast, older mice with genetic inactivation of GABA signaling by AgRP neurons had only transient reduction of feeding and body weight; their energy expenditure and glucose tolerance were unaffected. These results indicate that GABAergic signaling from AgRP neurons plays a key role in the control of feeding and metabolism through an age-dependent mechanism. This new genetic technique will augment current tools used to elucidate mechanisms underlying many physiological and neurological processes.
Collapse
|
40
|
Pugach EK, Richmond PA, Azofeifa JG, Dowell RD, Leinwand LA. Prolonged Cre expression driven by the α-myosin heavy chain promoter can be cardiotoxic. J Mol Cell Cardiol 2015; 86:54-61. [PMID: 26141530 PMCID: PMC4558343 DOI: 10.1016/j.yjmcc.2015.06.019] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/10/2015] [Accepted: 06/24/2015] [Indexed: 12/23/2022]
Abstract
Studying the importance of genetic factors in a desired cell type or tissue necessitates the use of precise genetic tools. With the introduction of bacteriophage Cre recombinase/loxP mediated DNA editing and promoter-specific Cre expression, it is feasible to generate conditional knockout mice in which particular genes are disrupted in a cell type-specific manner in vivo. In cardiac myocytes, this is often achieved through α-myosin heavy chain promoter (αMyHC)-driven Cre expression in conjunction with a loxP-site flanked gene of interest. Recent studies in other cell types demonstrate toxicity of Cre expression through induction of DNA damage. However, it is unclear to what extent the traditionally used αMyHC-Cre line [1] may exhibit cardiotoxicity. Further, the genotype of αMyHC-Cre(+/-) is not often included as a control group in cardiac myocyte-specific knockout studies. Here we present evidence that these αMyHC-Cre(+/-) mice show molecular signs of cardiac toxicity by 3months of age and exhibit decreased cardiac function by 6months of age compared to wild-type littermates. Hearts from αMyHC-Cre(+/-) mice also display evidence of fibrosis, inflammation, and DNA damage. Interestingly, some of the early functional changes observed in αMyHC-Cre(+/-) mice are sexually dimorphic. Given the high level of Cre recombinase expression resulting from expression from the αMyHC promoter, we asked if degenerate loxP-like sites naturally exist in the mouse genome and if so, whether they are affected by Cre in the absence of canonical loxP-sites. Using a novel bioinformatics search tool, we identified 619 loxP-like sites with 4 or less mismatches to the canonical loxP-site. 227 sites overlapped with annotated genes and 55 of these genes were expressed in cardiac muscle. Expression of ~26% of the 27 genes tested was disrupted in αMyHC-Cre(+/-) mice indicating potential targeting by Cre. Taken together, these results highlight both the importance of using αMyHC-Cre mice as controls in conditional knockout studies as well as the need for a less cardiotoxic Cre driver for the field.
Collapse
Affiliation(s)
- Emily K Pugach
- University of Colorado at Boulder, Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, Boulder, CO 80303 USA.
| | - Phillip A Richmond
- University of Colorado at Boulder, Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, Boulder, CO 80303 USA.
| | - Joseph G Azofeifa
- University of Colorado at Boulder, Department of Computer Science, Boulder, CO 80303 USA.
| | - Robin D Dowell
- University of Colorado at Boulder, Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, Boulder, CO 80303 USA.
| | - Leslie A Leinwand
- University of Colorado at Boulder, Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, Boulder, CO 80303 USA.
| |
Collapse
|
41
|
Liu G, Elrashidy RA, Xiao N, Kavran M, Huang Y, Tao M, Powell CT, Kim E, Sadeghi G, Mohamed HE, Daneshgari F. Bladder function in mice with inducible smooth muscle-specific deletion of the manganese superoxide dismutase gene. Am J Physiol Cell Physiol 2015; 309:C169-78. [PMID: 25948732 DOI: 10.1152/ajpcell.00046.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/02/2015] [Indexed: 11/22/2022]
Abstract
Manganese superoxide dismutase (MnSOD) is considered a critical component of the antioxidant systems that protect against oxidative damage. We are interested in the role of oxidative stress in bladder detrusor smooth muscle (SM) in different disease states. In this study, we generated an inducible, SM-specific Sod2(-/-) mouse model to investigate the effects of MnSOD depletion on the function of the bladder. We crossbred floxed Sod2 (Sod2(lox/lox)) mice with mice containing heterozygous knock-in of a gene encoding a tamoxifen-activated Cre recombinase in the SM22α promoter locus [SM-CreER(T2)(ki)(Cre/+)]. We obtained Sod2(lox/lox),SM-CreER(T2)(ki)(Cre/+) mice and injected 8-wk-old males with 4-hydroxytamoxifen to induce Cre-mediated excision of the floxed Sod2 allele. Twelve weeks later, SM-specific deletion of Sod2 and depletion of MnSOD were confirmed by polymerase chain reaction, immunoblotting, and immunohistochemistry. SM-specific Sod2(-/-) mice exhibited normal growth with no gross abnormalities. A significant increase in nitrotyrosine concentration was found in bladder SM tissue of SM-specific Sod2(-/-) mice compared with both wild-type mice and Sod2(+/+), SM-CreER(T2)(ki)(Cre/+) mice treated with 4-hydroxytamoxifen. Assessment of 24-h micturition in SM-specific Sod2(-/-) mice revealed significantly higher voiding frequency compared with both wild-type and SM-specific Cre controls. Conscious cystometry revealed significantly shorter intercontraction intervals and lower functional bladder capacity in SM-specific Sod2(-/-) mice compared with wild-type mice. This novel model can be used for exploring the mechanistic role of oxidative stress in organs rich in SM in different pathological conditions.
Collapse
Affiliation(s)
- Guiming Liu
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Rania A Elrashidy
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Faculty of Pharmacy, Biochemistry Department, Zagazig University, Zagazig, Egypt
| | - Nan Xiao
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China; and
| | - Michael Kavran
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Yexiang Huang
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Mingfang Tao
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - C Thomas Powell
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Edward Kim
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ghazal Sadeghi
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Hoda E Mohamed
- Faculty of Pharmacy, Biochemistry Department, Zagazig University, Zagazig, Egypt
| | - Firouz Daneshgari
- Department of Urology, Case Western Reserve University School of Medicine, Cleveland, Ohio;
| |
Collapse
|
42
|
Coleman JLJ, Brennan K, Ngo T, Balaji P, Graham RM, Smith NJ. Rapid Knockout and Reporter Mouse Line Generation and Breeding Colony Establishment Using EUCOMM Conditional-Ready Embryonic Stem Cells: A Case Study. Front Endocrinol (Lausanne) 2015; 6:105. [PMID: 26175717 PMCID: PMC4485191 DOI: 10.3389/fendo.2015.00105] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022] Open
Abstract
As little as a decade ago, generation of a single knockout mouse line was an expensive and time-consuming undertaking available to relatively few researchers. The International Knockout Mouse Consortium, established in 2007, has revolutionized the use of such models by creating an open-access repository of embryonic stem (ES) cells that, through sequential breeding with first FLP1 recombinase and then Cre recombinase transgenic mice, facilitates germline global or conditional deletion of almost every gene in the mouse genome. In this Case Study, we describe our experience using the repository to create mouse lines for a variety of experimental purposes. Specifically, we discuss the process of obtaining germline transmission of two European Conditional Mouse Mutagenesis Program (EUCOMM) "knockout-first" gene targeted constructs and the advantages and pitfalls of using this system. We then outline our breeding strategy and the outcomes of our efforts to generate global and conditional knockouts and reporter mice for the genes of interest. Line maintenance, removal of recombinase transgenes, and cryopreservation are also considered. Our approach led to the generation of heterozygous knockout mice within 6 months of commencing breeding to the founder mice. By describing our experiences with the EUCOMM ES cells and subsequent breeding steps, we hope to assist other researchers with the application of this valuable approach to generating versatile knockout mouse lines.
Collapse
Affiliation(s)
- James L. J. Coleman
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Karen Brennan
- BioCORE, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Tony Ngo
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Poornima Balaji
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Robert M. Graham
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
- *Correspondence: Nicola J. Smith and Robert M. Graham, Victor Chang Cardiac Research Institute, Lowy Packer Building, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia, ;
| | - Nicola J. Smith
- Molecular Cardiology Program, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
- *Correspondence: Nicola J. Smith and Robert M. Graham, Victor Chang Cardiac Research Institute, Lowy Packer Building, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia, ;
| |
Collapse
|
43
|
Giusti SA, Vercelli CA, Vogl AM, Kolarz AW, Pino NS, Deussing JM, Refojo D. Behavioral phenotyping of Nestin-Cre mice: implications for genetic mouse models of psychiatric disorders. J Psychiatr Res 2014; 55:87-95. [PMID: 24768109 DOI: 10.1016/j.jpsychires.2014.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 11/19/2022]
Abstract
Genetic mouse models based on the Cre-loxP system have been extensively used to explore the influence of specific gene deletions on different aspects of behavioral neurobiology. However, the interpretation of the effects attributed to the gene deletion might be obscured by potential side effects secondary to the Cre recombinase transgene insertion or Cre activity, usually neither controlled nor reported. Here, we performed a comprehensive behavioral analysis of endophenotypes of neuropsychiatric disorders in the extensively used Nestin(Cre) mouse line, commonly employed to restrict genetic modifications to the CNS. We observed no alterations in locomotion, general exploratory activity, learning and memory, sociability, startle response and sensorimotor gating. Although the overall response to stimuli triggering anxiety-like behaviors remained unaltered in Nestin(Cre) mice, a strong impairment in the acquisition of both contextual- and cued-conditioned fear was observed. These results underline the importance of adequately controlling the behavioral performance of the employed Cre-lines per-se in pre-clinical neurobehavioral research.
Collapse
Affiliation(s)
- Sebastian A Giusti
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Claudia A Vercelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Annette M Vogl
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Adam W Kolarz
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natalia S Pino
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Damian Refojo
- Molecular Neurobiology, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
44
|
Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J Immunol Methods 2014; 408:89-100. [PMID: 24857755 DOI: 10.1016/j.jim.2014.05.009] [Citation(s) in RCA: 382] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 12/22/2022]
Abstract
Since the first example of conditional gene targeting in mice in 1994, the use of Cre recombinase and loxP flanked sequences has become an invaluable technique to generate tissue and temporal specific gene knockouts. The number of mouse strains expressing floxed-gene sequences, and tissue-specific or temporal-specific Cre-recombinase that have been reported in the literature has grown exponentially. However, increased use of this technology has highlighted several problems that can impact the interpretation of any phenotype observed in these mouse models. In particular, accurate knowledge of the specificity of Cre expression in each strain is critical in order to make conclusions about the role of specific cell types in the phenotypes observed. Cre-mediated deletion specificity and efficiency have been described in many different ways in the literature, making direct comparisons between these Cre strains impossible. Here we report crossing thirteen different myeloid-Cre mouse strains to ROSA-EYFP reporter mice and assaying YFP expression in a variety of naïve unstimulated hematopoietic cells, in parallel. By focusing on myeloid subsets, we directly compare the relative efficiency and specificity of myeloid deletion in these strains under steady-state conditions.
Collapse
|
45
|
Janbandhu VC, Moik D, Fässler R. Cre recombinase induces DNA damage and tetraploidy in the absence of loxP sites. Cell Cycle 2013; 13:462-70. [PMID: 24280829 DOI: 10.4161/cc.27271] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The spatiotemporal manipulations of gene expression by the Cre recombinase (Cre) of bacteriophage P1 has become an essential asset to understanding mammalian genetics. Accumulating evidence suggests that Cre activity can, in addition to excising targeted loxP sites, induce cytotoxic effects, including abnormal cell cycle progression, genomic instability, and apoptosis, which can accelerate cancer progression. It is speculated that these defects are caused by Cre-induced DNA damage at off-target sites. Here we report the formation of tetraploid keratinocytes in the epidermis of keratin 5 and/or keratin 14 promoter-driven Cre (KRT5- and KRT14-Cre) expressing mouse skin. Biochemical analyses and flow cytometry demonstrated that Cre expression also induces DNA damage, genomic instability, and tetraploidy in HCT116 cells, and live-cell imaging revealed an extension of the G 2 cell cycle phase followed by defective or skipping of mitosis as cause for the tetraploidy. Since tetraploidy eventually leads to aneuploidy, a hallmark of cancer, our findings highlight the importance of distinguishing non-specific cytopathic effects from specific Cre/loxP-driven genetic manipulations when using Cre-mediated gene deletions.
Collapse
Affiliation(s)
- Vaibhao C Janbandhu
- Max-Planck-Institute of Biochemistry; Department of Molecular Medicine; Martinsried, Germany
| | - Daniel Moik
- Max-Planck-Institute of Biochemistry; Department of Molecular Medicine; Martinsried, Germany
| | - Reinhard Fässler
- Max-Planck-Institute of Biochemistry; Department of Molecular Medicine; Martinsried, Germany
| |
Collapse
|
46
|
Nookaew I, Thorell K, Worah K, Wang S, Hibberd ML, Sjövall H, Pettersson S, Nielsen J, Lundin SB. Transcriptome signatures in Helicobacter pylori-infected mucosa identifies acidic mammalian chitinase loss as a corpus atrophy marker. BMC Med Genomics 2013; 6:41. [PMID: 24119614 PMCID: PMC4015281 DOI: 10.1186/1755-8794-6-41] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 10/01/2013] [Indexed: 12/20/2022] Open
Abstract
Background The majority of gastric cancer cases are believed to be caused by chronic infection with the bacterium Helicobacter pylori, and atrophic corpus gastritis is a predisposing condition to gastric cancer development. We aimed to increase understanding of the molecular details of atrophy by performing a global transcriptome analysis of stomach tissue. Methods Biopsies from patients with different stages of H. pylori infection were taken from both the antrum and corpus mucosa and analyzed on microarrays. The stages included patients without current H. pylori infection, H. pylori-infected without corpus atrophy and patients with current or past H. pylori-infection with corpus-predominant atrophic gastritis. Results Using clustering and integrated analysis, we found firm evidence for antralization of the corpus mucosa of atrophy patients. This antralization harbored gain of gastrin expression, as well as loss of expression of corpus-related genes, such as genes associated with acid production, energy metabolism and blood clotting. The analyses provided detailed molecular evidence for simultaneous intestinal metaplasia (IM) and spasmolytic polypeptide expressing metaplasia (SPEM) in atrophic corpus tissue. Finally, acidic mammalian chitinase, a chitin-degrading enzyme produced by chief cells, was shown to be strongly down-regulated in corpus atrophy. Conclusions Transcriptome analysis revealed several gene groups which are related to development of corpus atrophy, some of which were increased also in H. pylori-infected non-atrophic patients. Furthermore, loss of acidic chitinase expression is a promising marker for corpus atrophy.
Collapse
Affiliation(s)
- Intawat Nookaew
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Deussing JM. Targeted mutagenesis tools for modelling psychiatric disorders. Cell Tissue Res 2013; 354:9-25. [PMID: 24078022 DOI: 10.1007/s00441-013-1708-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/16/2013] [Indexed: 12/15/2022]
Abstract
In the 1980s, the basic principles of gene targeting were discovered and forged into sharp tools for efficient and precise engineering of the mouse genome. Since then, genetic mouse models have substantially contributed to our understanding of major neurobiological concepts and are of utmost importance for our comprehension of neuropsychiatric disorders. The "domestication" of site-specific recombinases and the continuous creative technological developments involving the implementation of previously identified biological principles such as transcriptional and posttranslational control now enable conditional mutagenesis with high spatial and temporal resolution. The initiation and successful accomplishment of large-scale efforts to annotate functionally the entire mouse genome and to build strategic resources for the research community have significantly accelerated the rapid proliferation and broad propagation of mouse genetic tools. Addressing neurobiological processes with the assistance of genetic mouse models is a routine procedure in psychiatric research and will be further extended in order to improve our understanding of disease mechanisms. In light of the highly complex nature of psychiatric disorders and the current lack of strong causal genetic variants, a major future challenge is to model of psychiatric disorders more appropriately. Humanized mice, and the recently developed toolbox of site-specific nucleases for more efficient and simplified tailoring of the genome, offer the perspective of significantly improved models. Ultimately, these tools will push the limits of gene targeting beyond the mouse to allow genome engineering in any model organism of interest.
Collapse
Affiliation(s)
- Jan M Deussing
- Max Planck Institute of Psychiatry, Molecular Neurogenetics, Kraepelinstrasse 2-10, 80804, Munich, Germany,
| |
Collapse
|
48
|
Capoccia BJ, Jin RU, Kong YY, Peek RM, Fassan M, Rugge M, Mills JC. The ubiquitin ligase Mindbomb 1 coordinates gastrointestinal secretory cell maturation. J Clin Invest 2013; 123:1475-1491. [PMID: 23478405 PMCID: PMC3613919 DOI: 10.1172/jci65703] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 01/17/2013] [Indexed: 01/04/2023] Open
Abstract
After cell fate specification, differentiating cells must amplify the specific subcellular features required for their specialized function. How cells regulate such subcellular scaling is a fundamental unanswered question. Here, we show that the E3 ubiquitin ligase Mindbomb 1 (MIB1) is required for the apical secretory apparatus established by gastric zymogenic cells as they differentiate from their progenitors. When Mib1 was deleted, death-associated protein kinase-1 (DAPK1) was rerouted to the cell base, microtubule-associated protein 1B (MAP1B) was dephosphorylated, and the apical vesicles that normally support mature secretory granules were dispersed. Consequently, secretory granules did not mature. The transcription factor MIST1 bound the first intron of Mib1 and regulated its expression. We further showed that loss of MIB1 and dismantling of the apical secretory apparatus was the earliest quantifiable aberration in zymogenic cells undergoing transition to a precancerous metaplastic state in mouse and human stomach. Our results reveal a mechanistic pathway by which cells can scale up a specific, specialized subcellular compartment to alter function during differentiation and scale it down during disease.
Collapse
Affiliation(s)
- Benjamin J. Capoccia
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Ramon U. Jin
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Young-Yun Kong
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Richard M. Peek
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Matteo Fassan
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Massimo Rugge
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Jason C. Mills
- Division of Gastroenterology, Departments of Medicine, Developmental Biology, and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
Pathology and Cytopathology Unit, Department of Medicine, University of Padua, Padua, Italy
| |
Collapse
|
49
|
Eroshenko N, Church GM. Mutants of Cre recombinase with improved accuracy. Nat Commun 2013; 4:2509. [PMID: 24056590 PMCID: PMC3972015 DOI: 10.1038/ncomms3509] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 08/27/2013] [Indexed: 01/17/2023] Open
Abstract
Despite rapid advances in genome engineering technologies, inserting genes into precise locations in the human genome remains an outstanding problem. It has been suggested that site-specific recombinases can be adapted towards use as transgene delivery vectors. The specificity of recombinases can be altered either with directed evolution or via fusions to modular DNA-binding domains. Unfortunately, both wild-type and altered variants often have detectable activities at off-target sites. Here we use bacterial selections to identify mutations in the dimerization surface of Cre recombinase (R32V, R32M and 303GVSdup) that improve the accuracy of recombination. The mutants are functional in bacteria, in human cells and in vitro (except for 303GVSdup, which we did not purify), and have improved selectivity against both model off-target sites and the entire E. coli genome. We propose that destabilizing binding cooperativity may be a general strategy for improving the accuracy of dimeric DNA-binding proteins.
Collapse
Affiliation(s)
- Nikolai Eroshenko
- Harvard School of Engineering and Applied Sciences, Cambridge, MA 02138, USA
| | | |
Collapse
|
50
|
Abstract
Eradication of Helicobacter pylori correlates with regeneration of the gastric epithelium, ulcer healing and re-expression of the gastric morphogen Sonic Hedgehog (Shh). We sought to identify the role of Shh as a regulator of gastric epithelial regeneration during wound healing. A mouse model expressing a parietal cell-specific, tamoxifen-inducible deletion of Shh (HKCre(ERT2);Shh(flox/flox) or PC-iShhKO) was developed. Stomachs were collected and compared 7-150 days after the final vehicle or tamoxifen injection. Ulcers were induced in both controls and PC-iShhKO mice using acetic acid and ulcer size compared 1 and 7 days post induction. (1) Re-expression of Shh correlates with decreased hyperproliferation: Compared to controls, PC-iShhKO mice developed foveolar hyperplasia. Restoration of normal gastric epithelial architecture and differentiation correlated with the re-expression of Shh in PC-iShhKO mice 150 days after the final tamoxifen injection. At the tamoxifen dose used to induce Cre recombination there was no genotoxicity reported in either HKCre(ERT2) or Shh(flox/flox) control mouse stomachs. (2) Delayed wound healing in PC-iShhKO mouse stomachs: To identify the role of Shh in gastric regeneration, an acetic acid ulcer was induced in control and PC-iShhKO mice. Ulcers began to heal in control mice by 7 days after induction. Ulcer healing was documented by decreased ulcer size, angiogenesis, macrophage infiltration and formation of granulation tissue that correlated with the re-expression of Shh within the ulcerated tissue. PC-iShhKO mice did not show evidence of ulcer healing. Re-expression of Shh contributes to gastric regeneration. Our current study may have clinical implications given that eradication of H. pylori correlates with re-expression of Shh, regeneration of the gastric epithelium and ulcer healing.
Collapse
|