101
|
Abstract
Lineage tracing allows for identification of all progeny produced by a single cell or groups of cells and can thus be used to assess developmental fate of cells. Here we focus on one of the most widely used lineage tracing approaches that utilize the Cre/loxP system for site-specific genetic recombination in studying the developmental origins of lymphatic endothelial cells (LECs) in the mouse embryo. We discuss general considerations for a successful Cre/loxP based lineage tracing experiment and provide information about strains that are available for genetic lineage tracing of LECs. A protocol for lineage tracing analysis of the lymphatic vasculature by whole-mount immunofluorescence in two embryonic tissues, the skin and the mesentery, is also provided.
Collapse
|
102
|
Multiple enhancer regions govern the transcription of CCN2 during embryonic development. J Cell Commun Signal 2017; 12:231-243. [PMID: 29256171 PMCID: PMC5842200 DOI: 10.1007/s12079-017-0440-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/05/2017] [Indexed: 01/22/2023] Open
Abstract
CCN2 is a critical matricellular protein that is expressed in several cells with major implications in physiology and different pathologies. However, the transcriptional regulation of this gene remains obscure. We used the Encyclopaedia of DNA Elements browser (ENCODE) to visualise the region spanning from 300 kb upstream to the CCN2 start site in silico in order to identify enhancer regions that regulate transcription of this gene. Selection was based on three criteria associated with enhancer regions: 1) H3K4me1 and H3K27ac histone modifications, 2) DNase I hypersensitivity of chromatin and 3) inter-species conservation. Reporter constructs were created with sequences spanning each of the regions of interest placed upstream of an Hsp68 silent proximal promoter sequence in order to drive the expression of β-galactosidase transgene. Each of these constructs was subsequently used to create transgenic mice in which reporter gene production was assessed at the E15.5 developmental stage. Four functional enhancers were identified, with each driving distinct, tissue-specific patterns of transgene expression. An enhancer located -100 kb from the CCN2 transcription start site facilitated expression within vascular tissue. An enhancer -135 kb upstream of CCN2 drove expression within the articular chondrocytes of synovial joints. The other two enhancers, located at -198 kb and -229 kb, mediated transgene expression within dermal fibroblasts, however the most prevalent activity was found within hypertrophic chondrocytes and periosteal tissue, respectively. These findings suggest that the global expression of CCN2 during development results from the activity of several tissue-specific enhancer regions in addition to proximal regulatory elements that have previously been demonstrated to drive transcription of the gene during development.
Collapse
|
103
|
Inducible overexpression of endothelial proNGF as a mouse model to study microvascular dysfunction. Biochim Biophys Acta Mol Basis Dis 2017; 1864:746-757. [PMID: 29253516 DOI: 10.1016/j.bbadis.2017.12.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022]
Abstract
Impaired maturation of nerve growth factor precursor (proNGF) and its accumulation has been reported in several neurodegenerative diseases, myocardial infarction and diabetes. To elucidate the direct impact of proNGF accumulation identified the need to create a transgenic model that can express fully mutated cleavage-resistant proNGF. Using Cre-Lox technology, we developed an inducible endothelial-specific proNGF transgenic mouse (proNGFLoxp) that overexpresses GFP-conjugated cleavage-resistant proNGF123 when crossed with VE-cadherin-CreERT2 (Cre). Expression of proNGF, inflammatory mediators, NGF and VEGF was evaluated by PCR, Western blot and immunohistochemistry. EC-proNGF overexpression was confirmed using colocalization of anti-proNGF within retinal vasculature. EC-proNGF did not cause retinal neurotoxicity or marked glial activation at 4-weeks. Microvascular preparation from Cre-proNGF mice showed significant imbalance of proNGF/NGF ratio, enhanced expression of TNF-α and p75NTR, and tendency to impair TrkA phosphorylation compared to controls. EC-proNGF overexpression triggered mRNA expression of p75NTR and inflammatory mediators in both retina and renal cortex compared to controls. EC-proNGF expression induced vascular permeability including breakdown of BRB and albuminuria in the kidney without affecting VEGF level at 4-weeks. Histopathological changes were assessed after 8-weeks and the results showed that EC-proNGF triggered formation of occluded (acellular) capillaries, hall mark of retinal ischemia. EC-proNGF resulted in glomerular enlargement and kidney fibrosis, hall mark of renal dysfunction. We have successfully created an inducible mouse model that can dissect the contribution of autocrine direct action of cleavage-resistant proNGF on systemic microvascular abnormalities in both retina and kidney, major targets for microvascular complication.
Collapse
|
104
|
Rahman AA, Lai NK, Albright JE, Urquhart PE, Webb AR, Morrison BE. Nigral dopaminergic neuron replenishment in adult mice through VE-cadherin-expressing neural progenitor cells. Neural Regen Res 2017; 12:1865-1869. [PMID: 29239333 PMCID: PMC5745841 DOI: 10.4103/1673-5374.219050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The function of dopaminergic neurons in the substantia nigra is of central importance to the coordination of movement by the brain's basal ganglia circuitry. This is evidenced by the loss of these neurons, resulting in the cardinal motor deficits associated with Parkinson's disease. In order to fully understand the physiology of these key neurons and develop potential therapies for their loss, it is essential to determine if and how dopaminergic neurons are replenished in the adult brain. Recent work has presented evidence for adult neurogenesis of these neurons by Nestin+/Sox2– neural progenitor cells. We sought to further validate this finding and explore a potential atypical origin for these progenitor cells. Since neural progenitor cells have a proximal association with the vasculature of the brain and subsets of endothelial cells are Nestin+, we hypothesized that dopaminergic neural progenitors might share a common cell lineage. Therefore, we employed a VE-cadherin promoter-driven CREERT2:THlox/THlox transgenic mouse line to ablate the tyrosine hydroxylase gene from endothelial cells in adult animals. After 26 weeks, but not 13 weeks, following the genetic blockade of tyrosine hydroxylase expression in VE-cadherin+ cells, we observed a significant reduction in tyrosine hydroxylase+ neurons in the substantia nigra. The results from this genetic lineage tracing study suggest that dopaminergic neurons are replenished in adult mice by a VE-cadherin+ progenitor cell population potentially arising from an endothelial lineage.
Collapse
Affiliation(s)
- Abir A Rahman
- Department of Biological Sciences, Boise State University, Boise, ID; Biomolecular Ph.D. Program, Boise State University, Boise, ID, USA
| | - Nathan K Lai
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Joshua E Albright
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Paige E Urquhart
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Abby R Webb
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Brad E Morrison
- Department of Biological Sciences, Boise State University, Boise, ID; Biomolecular Ph.D. Program, Boise State University, Boise, ID; Department of Biological Sciences, Boise State University, Boise, ID, USA
| |
Collapse
|
105
|
Patel J, Baz B, Wong HY, Lee JS, Khosrotehrani K. Accelerated Endothelial to Mesenchymal Transition Increased Fibrosis via Deleting Notch Signaling in Wound Vasculature. J Invest Dermatol 2017; 138:1166-1175. [PMID: 29248546 DOI: 10.1016/j.jid.2017.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/22/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022]
Abstract
Skin wound healing in adults is characterized by a peak of angiogenesis followed by regression of the excessive vasculature in parallel with collagen deposition and fibrosis in the wound. We hypothesized that regressing vessels in healing wounds were in fact entering an endothelial to mesenchymal transition contributing to scarring. Using vascular-specific fate tracking (Cdh5-creERt2/ROSA-YFP mice), full-thickness excisional wounds were analyzed to reveal a time-dependent transition from endothelial phenotype characterized by vascular endothelial-cadherin, CD31, and CD34 toward a mesenchymal phenotype characterized by alpha-smooth muscle actin and fibroblast-specific protein 1 expression. We next conditionally ablated RBPJ in the vasculature (Rbpjfl/fl/Cdh5-creERt2ROSA-YFP) to evaluate the role of canonical Notch signaling in this process. Endothelial to mesenchymal transition was clearly accelerated after the loss of Notch signaling within the vasculature. The acceleration of endothelial to mesenchymal transition resulted in delayed wound healing, increased fibrosis, and extensive scar tissue formation, with the rapid loss of key endothelial genes and proteins and upregulation of mesenchymal protein expression (alpha-smooth muscle actin and fibroblast-specific protein 1) in vessels. Our findings here uncover a cellular contributor to skin wound scarring through the process of endothelial to mesenchymal transition in skin wounds and demonstrate the importance of Notch signaling in regulating this critical process during healing.
Collapse
Affiliation(s)
- Jatin Patel
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Betoul Baz
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Ho Yi Wong
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - James S Lee
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia; UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
106
|
Myosin IIa is critical for cAMP-mediated endothelial secretion of von Willebrand factor. Blood 2017; 131:686-698. [PMID: 29208598 DOI: 10.1182/blood-2017-08-802140] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/14/2017] [Indexed: 01/10/2023] Open
Abstract
Nonmuscle myosin II has been implicated in regulation of von Willebrand factor (VWF) release from endothelial Weibel-Palade bodies (WPBs), but the specific role of myosin IIa isoform is poorly defined. Here, we report that myosin IIa is expressed both in primary human endothelial cells and intact mouse vessels, essential for cyclic adenosine monophosphate (cAMP)-mediated endothelial VWF secretion. Downregulation of myosin IIa by shRNAs significantly suppressed both forskolin- and epinephrine-induced VWF secretion. Endothelium-specific myosin IIa knockout mice exhibited impaired epinephrine-stimulated VWF release, prolonged bleeding time, and thrombosis. Further study showed that in resting cells, myosin IIa deficiency disrupted the peripheral localization of Rab27-positive WPBs along stress fibers; on stimulation by cAMP agonists, myosin IIa in synergy with zyxin promotes the formation of a functional actin framework, which is derived from preexisting cortical actin filaments, around WPBs, facilitating fusion and subsequent exocytosis. In summary, our findings not only identify new functions of myosin IIa in regulation of WPB positioning and the interaction between preexisting cortical actin filaments and exocytosing vesicles before fusion but also reveal myosin IIa as a physiological regulator of endothelial VWF secretion in stress-induced hemostasis and thrombosis.
Collapse
|
107
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 571] [Impact Index Per Article: 81.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
108
|
Cain-Hom C, Splinter E, van Min M, Simonis M, van de Heijning M, Martinez M, Asghari V, Cox JC, Warming S. Efficient mapping of transgene integration sites and local structural changes in Cre transgenic mice using targeted locus amplification. Nucleic Acids Res 2017; 45:e62. [PMID: 28053125 PMCID: PMC5416772 DOI: 10.1093/nar/gkw1329] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/20/2016] [Indexed: 12/23/2022] Open
Abstract
Cre/LoxP technology is widely used in the field of mouse genetics for spatial and/or temporal regulation of gene function. For Cre lines generated via pronuclear microinjection of a Cre transgene construct, the integration site is random and in most cases not known. Integration of a transgene can disrupt an endogenous gene, potentially interfering with interpretation of the phenotype. In addition, knowledge of where the transgene is integrated is important for planning of crosses between animals carrying a conditional allele and a given Cre allele in case the alleles are on the same chromosome. We have used targeted locus amplification (TLA) to efficiently map the transgene location in seven previously published Cre and CreERT2 transgenic lines. In all lines, transgene insertion was associated with structural changes of variable complexity, illustrating the importance of testing for rearrangements around the integration site. In all seven lines the exact integration site and breakpoint sequences were identified. Our methods, data and genotyping assays can be used as a resource for the mouse community and our results illustrate the power of the TLA method to not only efficiently map the integration site of any transgene, but also provide additional information regarding the transgene integration events.
Collapse
Affiliation(s)
- Carol Cain-Hom
- Department of Transgenic Technology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Erik Splinter
- Cergentis BV, Yalelaan 62, 3584 CM Utrecht, the Netherlands
| | - Max van Min
- Cergentis BV, Yalelaan 62, 3584 CM Utrecht, the Netherlands
| | | | | | - Maria Martinez
- Department of Transgenic Technology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vida Asghari
- Department of Transgenic Technology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - J Colin Cox
- Department of Transgenic Technology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Søren Warming
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
109
|
Low-dose tamoxifen treatment in juvenile males has long-term adverse effects on the reproductive system: implications for inducible transgenics. Sci Rep 2017; 7:8991. [PMID: 28827578 PMCID: PMC5566418 DOI: 10.1038/s41598-017-09016-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/17/2017] [Indexed: 11/08/2022] Open
Abstract
The tamoxifen-inducible Cre system is a popular transgenic method for controlling the induction of recombination by Cre at a specific time and in a specific cell type. However, tamoxifen is not an inert inducer of recombination, but an established endocrine disruptor with mixed agonist/antagonist activity acting via endogenous estrogen receptors. Such potentially confounding effects should be controlled for, but >40% of publications that have used tamoxifen to generate conditional knockouts have not reported even the minimum appropriate controls. To highlight the importance of this issue, the present study investigated the long-term impacts of different doses of a single systemic tamoxifen injection on the testis and the wider endocrine system. We found that a single dose of tamoxifen less than 10% of the mean dose used for recombination induction, caused adverse effects to the testis and to the reproductive endocrine system that persisted long-term. These data raise significant concerns about the widespread use of tamoxifen induction of recombination, and highlight the importance of including appropriate controls in all pathophysiological studies using this means of induction.
Collapse
|
110
|
Schlecht A, Leimbeck SV, Jägle H, Feuchtinger A, Tamm ER, Braunger BM. Deletion of Endothelial Transforming Growth Factor-β Signaling Leads to Choroidal Neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2570-2589. [PMID: 28823871 DOI: 10.1016/j.ajpath.2017.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 12/13/2022]
Abstract
The molecular pathogenesis of choroidal neovascularization (CNV), an angiogenic process that critically contributes to vision loss in age-related macular degeneration, is unclear. Herein, we analyzed the role of transforming growth factor (TGF)-β signaling for CNV formation by generating a series of mutant mouse models with induced conditional deletion of TGF-β signaling in the entire eye, the retinal pigment epithelium (RPE), or the vascular endothelium. Deletion of TGF-β signaling in the eye caused CNV, irrespectively if it was ablated in newborn or 3-week-old mice. Areas of CNV showed photoreceptor degeneration, multilayered RPE, basal lamina deposits, and accumulations of monocytes/macrophages. The changes progressed, leading to marked structural and functional alterations of the retina. Although the specific deletion of TGF-β signaling in the RPE caused no obvious changes, specific deletion in vascular endothelial cells caused CNV and a phenotype similar to that observed after the deletion in the entire eye. We conclude that impairment of TGF-β signaling in the vascular endothelium of the eye is sufficient to trigger CNV formation. Our findings highlight the importance of TGF-β signaling as a key player in the development of ocular neovascularization and indicate a fundamental role of TGF-β signaling in the pathogenesis of age-related macular degeneration.
Collapse
Affiliation(s)
- Anja Schlecht
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Sarah V Leimbeck
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University Clinic Regensburg, Regensburg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum Munich, Munich, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| | - Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
111
|
Toullec A, Buard V, Rannou E, Tarlet G, Guipaud O, Robine S, Iruela-Arispe ML, François A, Milliat F. HIF-1α Deletion in the Endothelium, but Not in the Epithelium, Protects From Radiation-Induced Enteritis. Cell Mol Gastroenterol Hepatol 2017; 5:15-30. [PMID: 29276749 PMCID: PMC5738457 DOI: 10.1016/j.jcmgh.2017.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/08/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Radiation therapy in the pelvic area is associated with side effects that impact the quality of life of cancer survivors. Interestingly, the gastrointestinal tract is able to adapt to significant changes in oxygen availability, suggesting that mechanisms related to hypoxia sensing help preserve tissue integrity in this organ. However, hypoxia-inducible factor (HIF)-dependent responses to radiation-induced gut toxicity are unknown. Radiation-induced intestinal toxicity is a complex process involving multiple cellular compartments. Here, we investigated whether epithelial or endothelial tissue-specific HIF-1α deletion could affect acute intestinal response to radiation. METHODS Using constitutive and inducible epithelial or endothelial tissue-specific HIF-1α deletion, we evaluated the consequences of epithelial or endothelial HIF-1α deletion on radiation-induced enteritis after localized irradiation. Survival, radiation-induced tissue injury, molecular inflammatory profile, tissue hypoxia, and vascular injury were monitored. RESULTS Surprisingly, epithelium-specific HIF-1α deletion does not alter radiation-induced intestinal injury. However, irradiated VECad-Cre+/-HIF-1αFL/FL mice present with lower radiation-induced damage, showed a preserved vasculature, reduced hypoxia, and reduced proinflammatory response compared with irradiated HIF-1αFL/FL mice. CONCLUSIONS We demonstrate in vivo that HIF-1α impacts radiation-induced enteritis and that this role differs according to the targeted cell type. Our work provides a new role for HIF-1α and endothelium-dependent mechanisms driving inflammatory processes in gut mucosae. Results presented show that effects on normal tissues have to be taken into account in approaches aiming to modulate hypoxia or hypoxia-related molecular mechanisms.
Collapse
Key Words
- EndoMT, endothelial-to-mesenchymal transition
- Endothelium
- HIF, hypoxia-inducible factor
- HIF-1α
- HIF-1αFl/FL, HIF-1α floxed mice
- HIMEC, human intestinal microvascular endothelial cells
- HUVEC, human umbilical vein endothelial cells
- IL, interleukin
- PAI-1, plasminogen activator inhibitor type-1
- PCR, polymerase chain reaction
- ROSA, ROSA26R LacZ reporter mice
- Radiation
- Sham-IR, sham-irradiation
- TBI, total body irradiation
- VECad-Cre, VE-cadherin-Cre mice
Collapse
Affiliation(s)
- Aurore Toullec
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Valérie Buard
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Emilie Rannou
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California
| | - Georges Tarlet
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | | | - M. Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California
| | - Agnès François
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
- Correspondence Address correspondence to: Fabien Milliat, PhD, Research Laboratory of Radiobiology and Radiopathology, Institute for Radiological Protection and Nuclear Safety, 92265 Fontenay-aux-Roses, France.Research Laboratory of Radiobiology and RadiopathologyInstitute for Radiological Protection and Nuclear Safety92265 Fontenay-aux-RosesFrance
| |
Collapse
|
112
|
Endothelial cell-specific activation of transforming growth factor-β signaling in mice induces cutaneous, visceral, and microvascular fibrosis. J Transl Med 2017; 97:806-818. [PMID: 28346399 PMCID: PMC6530474 DOI: 10.1038/labinvest.2017.23] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023] Open
Abstract
In this study, we tested the hypothesis that constitutive endothelial cell-specific activation of TGF-β signaling induces tissue fibrosis and vasculopathy resembling the characteristic fibrotic and vascular alterations of systemic sclerosis. Transgenic mice with inducible expression of a constitutively active TGF-β receptor I specifically in endothelial cells were generated by intercrossing mice harboring a constitutively active TGF-β receptor I with a mouse strain containing the endothelial cell-specific Cdh5 gene promoter directing the tamoxifen-inducible expression of the Cre-ERT2 cassette. Administration of tamoxifen to these mice would result in constitutive TGF-β activation and signaling confined to endothelial lineage cells. The effects of constitutive TGF-β endothelial cell activation were assessed by histopathological examination of skin and various internal organs, tissue hydroxyproline analysis, and assessment of expression of myofibroblast differentiation and TGF-β signaling genes employing real-time PCR and immunohistochemical staining of lung vessels for endothelial- and myofibroblast-specific proteins. Constitutive TGFβ-1 signaling in endothelial cells resulted in cutaneous and visceral fibrosis with prominent fibrotic involvement of the lungs and severe perivascular and subendothelial fibrosis of small arterioles. A marked increase in the expression of fibrosis-associated genes and of genes indicative of myofibroblast activation was also found. Confocal microscopy of lung vessels showed evidence consistent with the induction of endothelial-to-mesenchymal transition (EndoMT). Taken together, our data indicate that transgenic mice with constitutive endothelial cell-specific activation of TGF-β signaling display severe cutaneous, pulmonary, and microvascular fibrosis resembling the fibrotic and microvascular alterations characteristic of systemic sclerosis.
Collapse
|
113
|
Kim B, Li J, Jang C, Arany Z. Glutamine fuels proliferation but not migration of endothelial cells. EMBO J 2017; 36:2321-2333. [PMID: 28659379 DOI: 10.15252/embj.201796436] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/14/2022] Open
Abstract
Endothelial metabolism is a key regulator of angiogenesis. Glutamine metabolism in endothelial cells (ECs) has been poorly studied. We used genetic modifications and 13C tracing approaches to define glutamine metabolism in these cells. Glutamine supplies the majority of carbons in the tricyclic acid (TCA) cycle of ECs and contributes to lipid biosynthesis via reductive carboxylation. EC-specific deletion in mice of glutaminase, the initial enzyme in glutamine catabolism, markedly blunts angiogenesis. In cell culture, glutamine deprivation or inhibition of glutaminase prevents EC proliferation, but does not prevent cell migration, which relies instead on aerobic glycolysis. Without glutamine catabolism, there is near complete loss of TCA intermediates, with no compensation from glucose-derived anaplerosis. Mechanistically, addition of exogenous alpha-ketoglutarate replenishes TCA intermediates and rescues cellular growth, but simultaneously unveils a requirement for Rac1-dependent macropinocytosis to provide non-essential amino acids, including asparagine. Together, these data outline the dependence of ECs on glutamine for cataplerotic processes; the need for glutamine as a nitrogen source for generation of biomass; and the distinct roles of glucose and glutamine in EC biology.
Collapse
Affiliation(s)
- Boa Kim
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jia Li
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Chemistry and Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zoltan Arany
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA .,Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
114
|
Oskolkova O, Gawlak G, Tian Y, Ke Y, Sarich N, Son S, Andreasson K, Bochkov VN, Birukova AA, Birukov KG. Prostaglandin E receptor-4 receptor mediates endothelial barrier-enhancing and anti-inflammatory effects of oxidized phospholipids. FASEB J 2017; 31:4187-4202. [PMID: 28572443 DOI: 10.1096/fj.201601232rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/08/2017] [Indexed: 01/08/2023]
Abstract
Unlike other agonists that cause transient endothelial cell (EC) response, the products of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (PAPC) oxidation that contain cyclopenthenone groups, which recapitulate prostaglandin-like structure, cause sustained enhancement of the pulmonary EC barrier. The mechanisms that drive the sustained effects by oxidized PAPC (OxPAPC) remain unexplored. On the basis of the structural similarity of isoprostanoid moieties that are present in full-length oxygenated PAPC species, we used an inhibitory approach to perform the screening of prostanoid receptors as potential candidates that mediate OxPAPC effects. Results show that only prostaglandin E receptor-4 (EP4) was involved and mediated the sustained phase of the barrier-enhancing effects of OxPAPC that are associated with the activation of Rac GTPase and its cytoskeletal targets. EC incubation with OxPAPC also induced EP4 mRNA expression in pulmonary ECs and lung tissue. EP4 knockdown using gene-specific small interfering RNA did not affect the rapid phase of OxPAPC-induced EC barrier enhancement or the protective effects against thrombin-induced EC permeability, but abolished the advanced barrier enhancement phase and suppressed the protective effects of OxPAPC against more sustained EC barrier dysfunction and cell inflammatory response caused by TNF-α. Endothelial-specific knockout of the EP4 receptor in mice attenuated the protective effect of intravenous OxPAPC administration in the model of acute lung injury caused by intratracheal injection of LPS. Taken together, these results demonstrate a novel role for prostaglandin receptor EP4 in the mediation of barrier-enhancing and anti-inflammatory effects caused by oxidized phospholipids.-Oskolkova, O., Gawlak, G., Tian, Y., Ke, Y., Sarich, N., Son, S., Andreasson, K., Bochkov, V. N., Birukova, A. A., Birukov, K. G. Prostaglandin E receptor-4 receptor mediates endothelial barrier-enhancing and anti-inflammatory effects of oxidized phospholipids.
Collapse
Affiliation(s)
- Olga Oskolkova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Grzegorz Gawlak
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yufeng Tian
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yunbo Ke
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Nicolene Sarich
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Sophia Son
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Valery N Bochkov
- Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Anna A Birukova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA;
| |
Collapse
|
115
|
Ohmura T, Tian Y, Sarich N, Ke Y, Meliton A, Shah AS, Andreasson K, Birukov KG, Birukova AA. Regulation of lung endothelial permeability and inflammatory responses by prostaglandin A2: role of EP4 receptor. Mol Biol Cell 2017; 28:1622-1635. [PMID: 28428256 PMCID: PMC5469606 DOI: 10.1091/mbc.e16-09-0639] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 03/27/2017] [Accepted: 04/13/2017] [Indexed: 12/02/2022] Open
Abstract
PGA2 exhibits anti-inflammatory and barrier-protective effects on endothelial cells and in two mouse models of acute lung injury. PGA2-induced cytoskeletal remodeling and suppression of the NFκB pathway is mediated by prostanoid receptor EP4. Endothelial-specific EP4 knockout abolishes PGA2-protective effects in vitro and in vivo. The role of prostaglandin A2 (PGA2) in modulation of vascular endothelial function is unknown. We investigated effects of PGA2 on pulmonary endothelial cell (EC) permeability and inflammatory activation and identified a receptor mediating these effects. PGA2 enhanced the EC barrier and protected against barrier dysfunction caused by vasoactive peptide thrombin and proinflammatory bacterial wall lipopolysaccharide (LPS). Receptor screening using pharmacological and molecular inhibitory approaches identified EP4 as a novel PGA2 receptor. EP4 mediated barrier-protective effects of PGA2 by activating Rap1/Rac1 GTPase and protein kinase A targets at cell adhesions and cytoskeleton: VE-cadherin, p120-catenin, ZO-1, cortactin, and VASP. PGA2 also suppressed LPS-induced inflammatory signaling by inhibiting the NFκB pathway and expression of EC adhesion molecules ICAM1 and VCAM1. These effects were abolished by pharmacological or molecular inhibition of EP4. In vivo, PGA2 was protective in two distinct models of acute lung injury (ALI): LPS-induced inflammatory injury and two-hit ALI caused by suboptimal mechanical ventilation and injection of thrombin receptor–activating peptide. These protective effects were abolished in mice with endothelial-specific EP4 knockout. The results suggest a novel role for the PGA2–EP4 axis in vascular EC protection that is critical for improvement of pathological states associated with increased vascular leakage and inflammation.
Collapse
Affiliation(s)
- Tomomi Ohmura
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Yufeng Tian
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Nicolene Sarich
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Yunbo Ke
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Angelo Meliton
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Alok S Shah
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Anna A Birukova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637
| |
Collapse
|
116
|
Endothelial transcription factor KLF2 negatively regulates liver regeneration via induction of activin A. Proc Natl Acad Sci U S A 2017; 114:3993-3998. [PMID: 28348240 DOI: 10.1073/pnas.1613392114] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells (ECs) not only are important for oxygen delivery but also act as a paracrine source for signals that determine the balance between tissue regeneration and fibrosis. Here we show that genetic inactivation of flow-induced transcription factor Krüppel-like factor 2 (KLF2) in ECs results in reduced liver damage and augmentation of hepatocyte proliferation after chronic liver injury by treatment with carbon tetrachloride (CCl4). Serum levels of GLDH3 and ALT were significantly reduced in CCl4-treated EC-specific KLF2-deficient mice. In contrast, transgenic overexpression of KLF2 in liver sinusoidal ECs reduced hepatocyte proliferation. KLF2 induced activin A expression and secretion from endothelial cells in vitro and in vivo, which inhibited hepatocyte proliferation. However, loss or gain of KLF2 expression did not change capillary density and liver fibrosis, but significantly affected hepatocyte proliferation. Taken together, the data demonstrate that KLF2 induces an antiproliferative secretome, including activin A, which attenuates liver regeneration.
Collapse
|
117
|
Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, Rautou PE. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol 2017; 66:212-227. [PMID: 27423426 DOI: 10.1016/j.jhep.2016.07.009] [Citation(s) in RCA: 599] [Impact Index Per Article: 85.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 12/13/2022]
Abstract
Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells representing the interface between blood cells on the one side and hepatocytes and hepatic stellate cells on the other side. LSECs represent a permeable barrier. Indeed, the association of 'fenestrae', absence of diaphragm and lack of basement membrane make them the most permeable endothelial cells of the mammalian body. They also have the highest endocytosis capacity of human cells. In physiological conditions, LSECs regulate hepatic vascular tone contributing to the maintenance of a low portal pressure despite the major changes in hepatic blood flow occurring during digestion. LSECs maintain hepatic stellate cell quiescence, thus inhibiting intrahepatic vasoconstriction and fibrosis development. In pathological conditions, LSECs play a key role in the initiation and progression of chronic liver diseases. Indeed, they become capillarized and lose their protective properties, and they promote angiogenesis and vasoconstriction. LSECs are implicated in liver regeneration following acute liver injury or partial hepatectomy since they renew from LSECs and/or LSEC progenitors, they sense changes in shear stress resulting from surgery, and they interact with platelets and inflammatory cells. LSECs also play a role in hepatocellular carcinoma development and progression, in ageing, and in liver lesions related to inflammation and infection. This review also presents a detailed analysis of the technical aspects relevant for LSEC analysis including the markers these cells express, the available cell lines and the transgenic mouse models. Finally, this review provides an overview of the strategies available for a specific targeting of LSECs.
Collapse
Affiliation(s)
- Johanne Poisson
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sara Lemoinne
- INSERM, UMRS 938, Centre de Recherche Saint-Antoine, Université Pierre et Marie Curie Paris 6, Paris, France; Service d'hépatologie, Hôpital Saint-Antoine, APHP, Paris, France
| | - Chantal Boulanger
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - François Durand
- Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; INSERM, UMR-1149, Centre de Recherche sur l'inflammation, Paris-Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France
| | - Richard Moreau
- Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; INSERM, UMR-1149, Centre de Recherche sur l'inflammation, Paris-Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France
| | - Dominique Valla
- Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; INSERM, UMR-1149, Centre de Recherche sur l'inflammation, Paris-Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France
| | - Pierre-Emmanuel Rautou
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; INSERM, UMR-1149, Centre de Recherche sur l'inflammation, Paris-Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France.
| |
Collapse
|
118
|
Dey D, Bagarova J, Hatsell SJ, Armstrong KA, Huang L, Ermann J, Vonner AJ, Shen Y, Mohedas AH, Lee A, Eekhoff EMW, van Schie A, Demay MB, Keller C, Wagers AJ, Economides AN, Yu PB. Two tissue-resident progenitor lineages drive distinct phenotypes of heterotopic ossification. Sci Transl Med 2016; 8:366ra163. [PMID: 27881824 PMCID: PMC6407419 DOI: 10.1126/scitranslmed.aaf1090] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/23/2016] [Indexed: 12/31/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP), a congenital heterotopic ossification (HO) syndrome caused by gain-of-function mutations of bone morphogenetic protein (BMP) type I receptor ACVR1, manifests with progressive ossification of skeletal muscles, tendons, ligaments, and joints. In this disease, HO can occur in discrete flares, often triggered by injury or inflammation, or may progress incrementally without identified triggers. Mice harboring an Acvr1R206H knock-in allele recapitulate the phenotypic spectrum of FOP, including injury-responsive intramuscular HO and spontaneous articular, tendon, and ligament ossification. The cells that drive HO in these diverse tissues can be compartmentalized into two lineages: an Scx+ tendon-derived progenitor that mediates endochondral HO of ligaments and joints without exogenous injury, and a muscle-resident interstitial Mx1+ population that mediates intramuscular, injury-dependent endochondral HO. Expression of Acvr1R206H in either lineage confers aberrant gain of BMP signaling and chondrogenic differentiation in response to activin A and gives rise to mutation-expressing hypertrophic chondrocytes in HO lesions. Compared to Acvr1R206H, expression of the man-made, ligand-independent ACVR1Q207D mutation accelerates and increases the penetrance of all observed phenotypes, but does not abrogate the need for antecedent injury in muscle HO, demonstrating the need for an injury factor in addition to enhanced BMP signaling. Both injury-dependent intramuscular and spontaneous ligament HO in Acvr1R206H knock-in mice were effectively controlled by the selective ACVR1 inhibitor LDN-212854. Thus, diverse phenotypes of HO found in FOP are rooted in cell-autonomous effects of dysregulated ACVR1 signaling in nonoverlapping tissue-resident progenitor pools that may be addressed by systemic therapy or by modulating injury-mediated factors involved in their local recruitment.
Collapse
Affiliation(s)
- Devaveena Dey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Jana Bagarova
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Sarah J Hatsell
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Kelli A Armstrong
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Lily Huang
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Joerg Ermann
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ashley J Vonner
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Yue Shen
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Agustin H Mohedas
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Arthur Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Elisabeth M W Eekhoff
- Departments of Internal Medicine, Endocrine Section, and Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, Amsterdam 1007 MB, Netherlands
| | - Annelies van Schie
- Departments of Internal Medicine, Endocrine Section, and Epidemiology and Biostatistics, VU University Medical Center, PO Box 7057, Amsterdam 1007 MB, Netherlands
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, 12655 SW Beaverdam Road-West, Beaverton, OR 97005, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Aris N Economides
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
- Regeneron Genetics Center, Tarrytown, NY 10591, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
119
|
Bianchi R, Russo E, Bachmann SB, Proulx ST, Sesartic M, Smaadahl N, Watson SP, Buckley CD, Halin C, Detmar M. Postnatal Deletion of Podoplanin in Lymphatic Endothelium Results in Blood Filling of the Lymphatic System and Impairs Dendritic Cell Migration to Lymph Nodes. Arterioscler Thromb Vasc Biol 2016; 37:108-117. [PMID: 27810998 DOI: 10.1161/atvbaha.116.308020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/23/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The lymphatic vascular system exerts major physiological functions in the transport of interstitial fluid from peripheral tissues back to the blood circulation and in the trafficking of immune cells to lymph nodes. Previous studies in global constitutive knockout mice for the lymphatic transmembrane molecule podoplanin reported perinatal lethality and a complex phenotype with lung abnormalities, cardiac defects, lymphedema, blood-filled lymphatic vessels, and lack of lymph node organization, reflecting the importance of podoplanin expression not only by the lymphatic endothelium but also by a variety of nonendothelial cell types. Therefore, we aimed to dissect the specific role of podoplanin expressed by adult lymphatic vessels. APPROACH AND RESULTS We generated an inducible, lymphatic-specific podoplanin knockout mouse model (PdpnΔLEC) and induced gene deletion postnatally. PdpnΔLEC mice were viable, and their lymphatic vessels appeared morphologically normal with unaltered fluid drainage function. Intriguingly, PdpnΔLEC mice had blood-filled lymph nodes and vessels, most frequently in the neck and axillary region, and displayed a blood-filled thoracic duct, suggestive of retrograde filling of blood from the blood circulation into the lymphatic system. Histological and fluorescence-activated cell sorter analyses revealed normal lymph node organization with the presence of erythrocytes within lymph node lymphatic vessels but not surrounding high endothelial venules. Moreover, fluorescein isothiocyanate painting experiments revealed reduced dendritic cell migration to lymph nodes in PdpnΔLEC mice. CONCLUSIONS These results reveal an important role of podoplanin expressed by lymphatic vessels in preventing postnatal blood filling of the lymphatic vascular system and in contributing to efficient dendritic cell migration to the lymph nodes.
Collapse
Affiliation(s)
- Roberta Bianchi
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Erica Russo
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Samia B Bachmann
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Steven T Proulx
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Marko Sesartic
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Nora Smaadahl
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Steve P Watson
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Christopher D Buckley
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Cornelia Halin
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom
| | - Michael Detmar
- From the Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland (R.B., E.R., S.B.B., S.T.P., M.S., N.S., C.H., M.D.); Centre for Cardiovascular Sciences, College of Medical and Dental Sciences (S.P.W.) and Rheumatology Research Group, Institute for Biomedical Research, College of Medical and Dental Sciences (C.D.B.), University of Birmingham, United Kingdom.
| |
Collapse
|
120
|
Dong L, Yu WM, Zheng H, Loh ML, Bunting ST, Pauly M, Huang G, Zhou M, Broxmeyer HE, Scadden DT, Qu CK. Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment. Nature 2016; 539:304-308. [PMID: 27783593 DOI: 10.1038/nature20131] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
Abstract
Germline activating mutations of the protein tyrosine phosphatase SHP2 (encoded by PTPN11), a positive regulator of the RAS signalling pathway, are found in 50% of patients with Noonan syndrome. These patients have an increased risk of developing leukaemia, especially juvenile myelomonocytic leukaemia (JMML), a childhood myeloproliferative neoplasm (MPN). Previous studies have demonstrated that mutations in Ptpn11 induce a JMML-like MPN through cell-autonomous mechanisms that are dependent on Shp2 catalytic activity. However, the effect of these mutations in the bone marrow microenvironment remains unclear. Here we report that Ptpn11 activating mutations in the mouse bone marrow microenvironment promote the development and progression of MPN through profound detrimental effects on haematopoietic stem cells (HSCs). Ptpn11 mutations in mesenchymal stem/progenitor cells and osteoprogenitors, but not in differentiated osteoblasts or endothelial cells, cause excessive production of the CC chemokine CCL3 (also known as MIP-1α), which recruits monocytes to the area in which HSCs also reside. Consequently, HSCs are hyperactivated by interleukin-1β and possibly other proinflammatory cytokines produced by monocytes, leading to exacerbated MPN and to donor-cell-derived MPN following stem cell transplantation. Remarkably, administration of CCL3 receptor antagonists effectively reverses MPN development induced by the Ptpn11-mutated bone marrow microenvironment. This study reveals the critical contribution of Ptpn11 mutations in the bone marrow microenvironment to leukaemogenesis and identifies CCL3 as a potential therapeutic target for controlling leukaemic progression in Noonan syndrome and for improving stem cell transplantation therapy in Noonan-syndrome-associated leukaemias.
Collapse
Affiliation(s)
- Lei Dong
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Wen-Mei Yu
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hong Zheng
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Mignon L Loh
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of California at San Francisco, San Francisco, California 94122, USA
| | - Silvia T Bunting
- Department of Pathology, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia 30322, USA
| | - Melinda Pauly
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Gang Huang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio 45229, USA
| | - Muxiang Zhou
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - David T Scadden
- Center for Regenerative Medicine and MGH Cancer Center, Massachusetts General Hospital, Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University, Boston, Massachusetts 02114, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Division of Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
121
|
Tamoxifen dosing for Cre-mediated recombination in experimental bronchopulmonary dysplasia. Transgenic Res 2016; 26:165-170. [PMID: 27730498 DOI: 10.1007/s11248-016-9987-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/29/2016] [Indexed: 10/20/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth characterized by blunted post-natal lung development. BPD can be modelled in mice by exposure of newborn mouse pups to elevated oxygen levels. Little is known about the mechanisms of perturbed lung development associated with BPD. The advent of transgenic mice, where genetic rearrangements can be induced in particular cell-types at particular time-points during organogenesis, have great potential to explore the pathogenic mechanisms at play during arrested lung development. Many inducible, conditional transgenic technologies available rely on the application of the estrogen-receptor modulator, tamoxifen. While tamoxifen is well-tolerated and has been widely employed in adult mice, or in healthy developing mice; tamoxifen is not well-tolerated in combination with hyperoxia, in the most widely-used mouse model of BPD. To address this, we set out to establish a safe and effective tamoxifen dosing regimen that can be used in newborn mouse pups subjected to injurious stimuli, such as exposure to elevated levels of environmental oxygen. Our data reveal that a single intraperitoneal dose of tamoxifen of 0.2 mg applied to newborn mouse pups in 10 μl Miglyol vehicle was adequate to successfully drive Cre recombinase-mediated genome rearrangements by the fifth day of life, in a murine model of BPD. The number of recombined cells was comparable to that observed in regular tamoxifen administration protocols. These findings will be useful to investigators where tamoxifen dosing is problematic in the background of injurious stimuli and mouse models of human and veterinary disease.
Collapse
|
122
|
Heo KS, Berk BC, Abe JI. Disturbed Flow-Induced Endothelial Proatherogenic Signaling Via Regulating Post-Translational Modifications and Epigenetic Events. Antioxid Redox Signal 2016; 25:435-50. [PMID: 26714841 PMCID: PMC5076483 DOI: 10.1089/ars.2015.6556] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/02/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Hemodynamic shear stress, the frictional force exerted onto the vascular endothelial cell (EC) surface, influences vascular EC functions. Atherosclerotic plaque formation in the endothelium is known to be site specific: disturbed blood flow (d-flow) formed at the lesser curvature of the aortic arch and branch points promotes plaque formation, and steady laminar flow (s-flow) at the greater curvature is atheroprotective. RECENT ADVANCES Post-translational modifications (PTMs), including phosphorylation and SUMOylation, and epigenetic events, including DNA methylation and histone modifications, provide a new perspective on the pathogenesis of atherosclerosis, elucidating how gene expression is altered by d-flow. Activation of PKCζ and p90RSK, SUMOylation of ERK5 and p53, and DNA hypermethylation are uniquely induced by d-flow, but not by s-flow. CRITICAL ISSUES Extensive cross talk has been observed among the phosphorylation, SUMOylation, acetylation, and methylation PTMs, as well as among epigenetic events along the cascade of d-flow-induced signaling, from the top (mechanosensory systems) to the bottom (epigenetic events). In addition, PKCζ activation plays a role in regulating SUMOylation-related enzymes of PIAS4, p90RSK activation plays a role in regulating SUMOylation-related enzymes of Sentrin/SUMO-specific protease (SENP)2, and DNA methyltransferase SUMOylation may play a role in d-flow signaling. FUTURE DIRECTIONS Although possible contributions of DNA events such as histone modification and the epigenetic and cytosolic events of PTMs in d-flow signaling have become clearer, determining the interplay of each PTM and epigenetic event will provide a new paradigm to elucidate the difference between d-flow and s-flow and lead to novel therapeutic interventions to inhibit plaque formation. Antioxid. Redox Signal. 25, 435-450.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bradford C. Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York
| | - Jun-ichi Abe
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
123
|
Yurdagul A, Orr AW. Blood Brothers: Hemodynamics and Cell-Matrix Interactions in Endothelial Function. Antioxid Redox Signal 2016; 25:415-34. [PMID: 26715135 PMCID: PMC5011636 DOI: 10.1089/ars.2015.6525] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/25/2015] [Accepted: 12/23/2015] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Alterations in endothelial function contribute to a variety of vascular diseases. In pathological conditions, the endothelium shows a reduced ability to regulate vasodilation (endothelial dysfunction) and a conversion toward a proinflammatory and leaky phenotype (endothelial activation). At the interface between the vessel wall and blood, the endothelium exists in a complex microenvironment and must translate changes in these environmental signals to alterations in vessel function. Mechanical stimulation and endothelial cell interactions with the vascular matrix, as well as a host of soluble factors, coordinately contribute to this dynamic regulation. RECENT ADVANCES Blood hemodynamics play an established role in the regulation of endothelial function. However, a growing body of work suggests that subendothelial matrix composition similarly and coordinately regulates endothelial cell phenotype such that blood flow affects matrix remodeling, which affects the endothelial response to flow. CRITICAL ISSUES Hemodynamics and soluble factors likely affect endothelial matrix remodeling through multiple mechanisms, including transforming growth factor β signaling and alterations in cell-matrix receptors, such as the integrins. Likewise, differential integrin signaling following matrix remodeling appears to regulate several key flow-induced responses, including nitric oxide production, regulation of oxidant stress, and activation of proinflammatory signaling and gene expression. Microvascular remodeling responses, such as angiogenesis and arteriogenesis, may also show coordinated regulation by flow and matrix. FUTURE DIRECTIONS Identifying the mechanisms regulating the dynamic interplay between hemodynamics and matrix remodeling and their contribution to the pathogenesis of cardiovascular disease remains an important research area with therapeutic implications across a variety of conditions. Antioxid. Redox Signal. 25, 415-434.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| | - A. Wayne Orr
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| |
Collapse
|
124
|
Pedrosa AR, Trindade A, Carvalho C, Graça J, Carvalho S, Peleteiro MC, Adams RH, Duarte A. Endothelial Jagged1 promotes solid tumor growth through both pro-angiogenic and angiocrine functions. Oncotarget 2016. [PMID: 26213336 PMCID: PMC4695194 DOI: 10.18632/oncotarget.4380] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is an essential process required for tumor growth and progression. The Notch signaling pathway has been identified as a key regulator of the neo-angiogenic process. Jagged-1 (Jag1) is a Notch ligand required for embryonic and retinal vascular development, which direct contribution to the regulation of tumor angiogenesis remains to be fully characterized. The current study addresses the role of endothelial Jagged1-mediated Notch signaling in the context of tumoral angiogenesis in two different mouse tumor models: subcutaneous Lewis Lung Carcinoma (LLC) tumor transplants and the autochthonous Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP). The role of endothelial Jagged1 in tumor growth and neo-angiogenesis was investigated with endothelial-specific Jag1 gain- and loss-of-function mouse mutants (eJag1OE and eJag1cKO). By modulating levels of endothelial Jag1, we observed that this ligand regulates tumor vessel density, branching, and perivascular maturation, thus affecting tumor vascular perfusion. The pro-angiogenic function is exerted by its ability to positively regulate levels of Vegfr-2 while negatively regulating Vegfr-1. Additionally, endothelial Jagged1 appears to exert an angiocrine function possibly by activating Notch3/Hey1 in tumor cells, promoting proliferation, survival and epithelial-to-mesenchymal transition (EMT), potentiating tumor development. These findings provide valuable mechanistic insights into the role of endothelial Jagged1 in promoting solid tumor development and support the notion that it may constitute a promising target for cancer therapy.
Collapse
Affiliation(s)
- Ana-Rita Pedrosa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Catarina Carvalho
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - José Graça
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Sandra Carvalho
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Maria C Peleteiro
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Muenster, Germany.,Faculty of Medicine, University of Muenster, Muenster, Germany
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), University of Lisbon, Lisbon, Portugal.,Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
125
|
Ochsenbein AM, Karaman S, Proulx ST, Berchtold M, Jurisic G, Stoeckli ET, Detmar M. Endothelial cell-derived semaphorin 3A inhibits filopodia formation by blood vascular tip cells. Development 2016; 143:589-94. [PMID: 26884395 DOI: 10.1242/dev.127670] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Vascular endothelial growth factor (VEGF)-A is a well-known major chemoattractant driver of angiogenesis--the formation of new blood vessels from pre-existing ones. However, the repellent factors that fine-tune this angiogenic process remain poorly characterized. We investigated the expression and functional role of endothelial cell-derived semaphorin 3A (Sema3A) in retinal angiogenesis, using genetic mouse models. We found Sema3a mRNA expression in the ganglion cell layer and the presence of Sema3A protein on larger blood vessels and at the growing front of blood vessels in neonatal retinas. The Sema3A receptors neuropilin-1 and plexin-A1 were expressed by retinal blood vessels. To study the endothelial cell-specific role of Sema3A, we generated endothelial cell-specific Sema3A knockout mouse strains by constitutive or inducible vascular endothelial cadherin-Cre-mediated gene disruption. We found that in neonatal retinas of these mice, both the number and the length of tip cell filopodia were significantly increased and the leading edge growth pattern was irregular. Retinal explant experiments showed that recombinant Sema3A significantly decreased VEGF-A-induced filopodia formation. Endothelial cell-specific knockout of Sema3A had no impact on blood vessel density or skin vascular leakage in adult mice. These findings indicate that endothelial cell-derived Sema3A exerts repelling functions on VEGF-A-induced tip cell filopodia and that a lack of this signaling cannot be rescued by paracrine sources of Sema3A.
Collapse
Affiliation(s)
- Alexandra M Ochsenbein
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Sinem Karaman
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Michaela Berchtold
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Giorgia Jurisic
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Esther T Stoeckli
- Institute of Molecular Life Sciences, University of Zurich, Zurich 8057, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
126
|
Höfer T, Busch K, Klapproth K, Rodewald HR. Fate Mapping and Quantitation of Hematopoiesis In Vivo. Annu Rev Immunol 2016; 34:449-78. [DOI: 10.1146/annurev-immunol-032414-112019] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Kay Klapproth
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| |
Collapse
|
127
|
Abstract
Myocardial fibrosis is a significant global health problem associated with nearly all forms of heart disease. Cardiac fibroblasts comprise an essential cell type in the heart that is responsible for the homeostasis of the extracellular matrix; however, upon injury, these cells transform to a myofibroblast phenotype and contribute to cardiac fibrosis. This remodeling involves pathological changes that include chamber dilation, cardiomyocyte hypertrophy and apoptosis, and ultimately leads to the progression to heart failure. Despite the critical importance of fibrosis in cardiovascular disease, our limited understanding of the cardiac fibroblast impedes the development of potential therapies that effectively target this cell type and its pathological contribution to disease progression. This review summarizes current knowledge regarding the origins and roles of fibroblasts, mediators and signaling pathways known to influence fibroblast function after myocardial injury, as well as novel therapeutic strategies under investigation to attenuate cardiac fibrosis.
Collapse
Affiliation(s)
- Joshua G Travers
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Fadia A Kamal
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Jeffrey Robbins
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Katherine E Yutzey
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH
| | - Burns C Blaxall
- From the Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, OH.
| |
Collapse
|
128
|
Martinez-Corral I, Stanczuk L, Frye M, Ulvmar MH, Diéguez-Hurtado R, Olmeda D, Makinen T, Ortega S. Vegfr3-CreER T2 mouse, a new genetic tool for targeting the lymphatic system. Angiogenesis 2016; 19:433-45. [DOI: 10.1007/s10456-016-9505-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 03/03/2016] [Indexed: 01/26/2023]
|
129
|
Kur E, Kim J, Tata A, Comin CH, Harrington KI, Costa LDF, Bentley K, Gu C. Temporal modulation of collective cell behavior controls vascular network topology. eLife 2016; 5. [PMID: 26910011 PMCID: PMC4811760 DOI: 10.7554/elife.13212] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/23/2016] [Indexed: 01/14/2023] Open
Abstract
Vascular network density determines the amount of oxygen and nutrients delivered to host tissues, but how the vast diversity of densities is generated is unknown. Reiterations of endothelial-tip-cell selection, sprout extension and anastomosis are the basis for vascular network generation, a process governed by the VEGF/Notch feedback loop. Here, we find that temporal regulation of this feedback loop, a previously unexplored dimension, is the key mechanism to determine vascular density. Iterating between computational modeling and in vivo live imaging, we demonstrate that the rate of tip-cell selection determines the length of linear sprout extension at the expense of branching, dictating network density. We provide the first example of a host tissue-derived signal (Semaphorin3E-Plexin-D1) that accelerates tip cell selection rate, yielding a dense network. We propose that temporal regulation of this critical, iterative aspect of network formation could be a general mechanism, and additional temporal regulators may exist to sculpt vascular topology. DOI:http://dx.doi.org/10.7554/eLife.13212.001 Many animals have a network of blood vessels that supplies oxygen and nutrients to every part of the body. Each organ contains a unique pattern of blood vessels; some have lots of densely packed vessels, while others have fewer vessels that are more widely spaced. New blood vessels typically form by sprouting from the side of pre-existing vessels. This involves the endothelial cells that line the inner wall of blood vessels moving outwards to create a sprout that is made up of ‘tip cells’ and ‘stalk cells’. Tip cells are found at the front of the growing vessels and encourage the formation of new sprouts, while the stalk cells trail behind and elongate the sprout. Two signaling pathways that involve two proteins called VEGF and Notch interact with each other to control which cells become tip cells and which become stalk cells. Cells with higher levels of VEGF signaling will become tip cells. These cells also activate Notch signaling, which in turn blocks VEGF signaling in their neighboring cells. This feedback mechanism enables a new sprout to form by forcing cells present around a newly formed tip cell to become stalk cells. However, it was still not understood how the different organs develop blood vessel networks with different densities. In 2011, researchers revealed that two other proteins, Semaphorin3E and its receptor Plexin-D1, are expressed in tip cells in the back of the eye in mice and control the VEGF/Notch signaling pathway. Now Kur et al. – including some of the researchers involved in the 2011 work – have used a combination of predictive computer simulations and experimental approaches to understand this interaction in more detail. The analysis showed that Semaphorin3E and Plexin-D1 speed up VEGF/Notch signaling, which causes new tip cells to form at a faster rate, and results in a more densely packed network of blood vessels. For example, in mice that lack Semaphorin3E and Plexin-D1, VEGF/Notch signaling was slower and new tip cells formed more slowly, which resulted in the blood vessel network at the back of the mice’s eyes being less dense. Kur et al. propose that different organs have different ‘molecular metronomes’ that control the pace of VEGF/Notch signaling. A fast acting metronome would yield a dense network, while a slower one would form a less dense network. This helps to explain how diverse densities of blood vessel networks are formed in different organs. This work may aid efforts to develop therapeutic approaches for controlling the development of new blood vessels in cancers and other diseases. DOI:http://dx.doi.org/10.7554/eLife.13212.002
Collapse
Affiliation(s)
- Esther Kur
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jiha Kim
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Aleksandra Tata
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Cesar H Comin
- Instituto de Física de São Carlos, University of Sao Paulo, Sao Carlos, Brazil
| | - Kyle I Harrington
- Center for Vascular Biology Research, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Luciano da F Costa
- Instituto de Física de São Carlos, University of Sao Paulo, Sao Carlos, Brazil
| | - Katie Bentley
- Center for Vascular Biology Research, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
130
|
Rasip1 is essential to blood vessel stability and angiogenic blood vessel growth. Angiogenesis 2016; 19:173-90. [PMID: 26897025 DOI: 10.1007/s10456-016-9498-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/03/2016] [Indexed: 01/09/2023]
Abstract
Cardiovascular function depends on patent, continuous and stable blood vessel formation by endothelial cells (ECs). Blood vessel development initiates by vasculogenesis, as ECs coalesce into linear aggregates and organize to form central lumens that allow blood flow. Molecular mechanisms underlying in vivo vascular 'tubulogenesis' are only beginning to be unraveled. We previously showed that the GTPase-interacting protein called Rasip1 is required for the formation of continuous vascular lumens in the early embryo. Rasip1(-/-) ECs exhibit loss of proper cell polarity and cell shape, disrupted localization of EC-EC junctions and defects in adhesion of ECs to extracellular matrix. In vitro studies showed that Rasip1 depletion in cultured ECs blocked tubulogenesis. Whether Rasip1 is required in blood vessels after their initial formation remained unclear. Here, we show that Rasip1 is essential for vessel formation and maintenance in the embryo, but not in quiescent adult vessels. Rasip1 is also required for angiogenesis in three models of blood vessel growth: in vitro matrix invasion, retinal blood vessel growth and directed in vivo angiogenesis assays. Rasip1 is thus necessary in growing embryonic blood vessels, postnatal angiogenic sprouting and remodeling, but is dispensable for maintenance of established blood vessels, making it a potential anti-angiogenic therapeutic target.
Collapse
|
131
|
Endothelial Dicer promotes atherosclerosis and vascular inflammation by miRNA-103-mediated suppression of KLF4. Nat Commun 2016; 7:10521. [PMID: 26837267 PMCID: PMC4742841 DOI: 10.1038/ncomms10521] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs regulate the maladaptation of endothelial cells (ECs) to naturally occurring disturbed blood flow at arterial bifurcations resulting in arterial inflammation and atherosclerosis in response to hyperlipidemic stress. Here, we show that reduced endothelial expression of the RNAse Dicer, which generates almost all mature miRNAs, decreases monocyte adhesion, endothelial C–X–C motif chemokine 1 (CXCL1) expression, atherosclerosis and the lesional macrophage content in apolipoprotein E knockout mice (Apoe−/−) after exposure to a high-fat diet. Endothelial Dicer deficiency reduces the expression of unstable miRNAs, such as miR-103, and promotes Krüppel-like factor 4 (KLF4)-dependent gene expression in murine atherosclerotic arteries. MiR-103 mediated suppression of KLF4 increases monocyte adhesion to ECs by enhancing nuclear factor-κB-dependent CXCL1 expression. Inhibiting the interaction between miR-103 and KLF4 reduces atherosclerosis, lesional macrophage accumulation and endothelial CXCL1 expression. Overall, our study suggests that Dicer promotes endothelial maladaptation and atherosclerosis in part by miR-103-mediated suppression of KLF4. The RNAse III endonuclease Dicer is crucial for processing of pre-miRNAs in health and disease. Here the authors show that endothelial Dicer promotes atherosclerosis by increasing miR-103 levels leading to suppression of the anti-inflammatory transcription factor KLF4, thus suggesting a novel approach to treat this disease.
Collapse
|
132
|
He L, Liu Q, Hu T, Huang X, Zhang H, Tian X, Yan Y, Wang L, Huang Y, Miquerol L, Wythe JD, Zhou B. Genetic lineage tracing discloses arteriogenesis as the main mechanism for collateral growth in the mouse heart. Cardiovasc Res 2016; 109:419-30. [PMID: 26768261 PMCID: PMC4752045 DOI: 10.1093/cvr/cvw005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022] Open
Abstract
Aims Capillary and arterial endothelial cells share many common molecular markers in both the neonatal and adult hearts. Herein, we aim to establish a genetic tool that distinguishes these two types of vessels in order to determine the cellular mechanism underlying collateral artery formation. Methods and results Using Apln-GFP and Apln-LacZ reporter mice, we demonstrate that APLN expression is enriched in coronary vascular endothelial cells. However, APLN expression is reduced in coronary arterial endothelial cells. Genetic lineage tracing, using an Apln-CreER mouse line, robustly labelled capillary endothelial cells, but not arterial endothelial cells. We leveraged this differential activity of Apln-CreER to study collateral artery formation following myocardial infarction (MI). In a neonatal heart MI model, we found that Apln-CreER-labelled capillary endothelial cells do not contribute to the large collateral arteries. Instead, these large collateral arteries mainly arise from pre-existing, infrequently labelled coronary arteries, indicative of arteriogenesis. Furthermore, in an adult heart MI model, Apln-CreER activity also distinguishes large and small diameter arteries from capillaries. Lineage tracing in this setting demonstrated that most large and small coronary arteries in the infarcted myocardium and border region are derived not from capillaries, but from pre-existing arteries. Conclusion Apln-CreER-mediated lineage tracing distinguishes capillaries from large arteries, in both the neonatal and adult hearts. Through genetic fate mapping, we demonstrate that pre-existing arteries, but not capillaries, extensively contribute to collateral artery formation following myocardial injury. These results suggest that arteriogenesis is the major mechanism underlying collateral vessel formation.
Collapse
Affiliation(s)
- Lingjuan He
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyuan Hu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiuzhen Huang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xueying Tian
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Yan
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lucile Miquerol
- Aix Marseille Universite, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Joshua D Wythe
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
133
|
Hartmann DA, Underly RG, Watson AN, Shih AY. A murine toolbox for imaging the neurovascular unit. Microcirculation 2015; 22:168-82. [PMID: 25352367 DOI: 10.1111/micc.12176] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/22/2014] [Indexed: 12/13/2022]
Abstract
The neurovascular unit (NVU) coordinates many essential functions in the brain including blood flow control, nutrient delivery, and maintenance of BBB integrity. These functions are the result of a cellular and molecular interplay that we are just beginning to understand. Cells of the NVU can now be investigated in the intact brain through the combined use of high-resolution in vivo imaging and non-invasive molecular tools to observe and manipulate cell function. Mouse lines that target transgene expression to cells of the NVU will be of great value in future work. However, a detailed evaluation of target cell specificity and expression pattern within the brain is required for many existing lines. The purpose of this review was to catalog mouse lines available to cerebrovascular biologists and to discuss their utility and limitations in future imaging studies.
Collapse
Affiliation(s)
- David A Hartmann
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | |
Collapse
|
134
|
Spadoni I, Zagato E, Bertocchi A, Paolinelli R, Hot E, Di Sabatino A, Caprioli F, Bottiglieri L, Oldani A, Viale G, Penna G, Dejana E, Rescigno M. A gut-vascular barrier controls the systemic dissemination of bacteria. Science 2015; 350:830-4. [PMID: 26564856 DOI: 10.1126/science.aad0135] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In healthy individuals, the intestinal microbiota cannot access the liver, spleen, or other peripheral tissues. Some pathogenic bacteria can reach these sites, however, and can induce a systemic immune response. How such compartmentalization is achieved is unknown. We identify a gut-vascular barrier (GVB) in mice and humans that controls the translocation of antigens into the blood stream and prohibits entry of the microbiota. Salmonella typhimurium can penetrate the GVB in a manner dependent on its pathogenicity island (Spi) 2-encoded type III secretion system and on decreased β-catenin-dependent signaling in gut endothelial cells. The GVB is modified in celiac disease patients with elevated serum transaminases, which indicates that GVB dismantling may be responsible for liver damage in these patients. Understanding the GVB may provide new insights into the regulation of the gut-liver axis.
Collapse
Affiliation(s)
- Ilaria Spadoni
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Elena Zagato
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Alice Bertocchi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Roberta Paolinelli
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Edina Hot
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Antonio Di Sabatino
- First Department of Medicine, St. Matteo Hospital, University of Pavia, Pavia, Italy
| | - Flavio Caprioli
- Unità Operativa Gastroenterologia ed Endoscopia, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico di Milano, and Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Milan, Italy
| | - Luca Bottiglieri
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Amanda Oldani
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Giuseppe Viale
- Department of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Giuseppe Penna
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Elisabetta Dejana
- The Italian Foundation for Cancer Research (FIRC) Institute of Molecular Oncology (IFOM), Milan, Italy. Department of Biosciences, Università degli Studi di Milano, Italy. Department of Genetics, Immunology and Pathology, Uppsala University, Uppsala, Sweden
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy. Department of Biosciences, Università degli Studi di Milano, Italy.
| |
Collapse
|
135
|
Endothelial Rictor is crucial for midgestational development and sustained and extensive FGF2-induced neovascularization in the adult. Sci Rep 2015; 5:17705. [PMID: 26635098 PMCID: PMC4669526 DOI: 10.1038/srep17705] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/03/2015] [Indexed: 12/14/2022] Open
Abstract
To explore the general requirement of endothelial mTORC2 during embryonic and
adolescent development, we knocked out the essential mTORC2 component Rictor
in the mouse endothelium in the embryo, during adolescence and in endothelial cells
in vitro. During embryonic development, Rictor knockout resulted
in growth retardation and lethality around embryonic day 12. We detected reduced
peripheral vascularization and delayed ossification of developing fingers, toes and
vertebrae during this confined midgestational period. Rictor knockout did not
affect viability, weight gain, and vascular development during further adolescence.
However during this period, Rictor knockout prevented skin capillaries to
gain larger and heterogeneously sized diameters and remodeling into tortuous vessels
in response to FGF2. Rictor knockout strongly reduced extensive FGF2-induced
neovascularization and prevented hemorrhage in FGF2-loaded matrigel plugs.
Rictor knockout also disabled the formation of capillary-like networks by
FGF2-stimulated mouse aortic endothelial cells in vitro. Low RICTOR
expression was detected in quiescent, confluent mouse aortic endothelial cells,
whereas high doses of FGF2 induced high RICTOR expression that was associated with
strong mTORC2-specific protein kinase Cα and AKT phosphorylation. We
demonstrate that the endothelial FGF-RICTOR axis is not required during endothelial
quiescence, but crucial for midgestational development and sustained and extensive
neovascularization in the adult.
Collapse
|
136
|
In vivo evidence for an endothelium-dependent mechanism in radiation-induced normal tissue injury. Sci Rep 2015; 5:15738. [PMID: 26510580 PMCID: PMC4625166 DOI: 10.1038/srep15738] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 09/29/2015] [Indexed: 11/30/2022] Open
Abstract
The pathophysiological mechanism involved in side effects of radiation therapy, and especially the role of the endothelium remains unclear. Previous results showed that plasminogen activator inhibitor-type 1 (PAI-1) contributes to radiation-induced intestinal injury and suggested that this role could be driven by an endothelium-dependent mechanism. We investigated whether endothelial-specific PAI-1 deletion could affect radiation-induced intestinal injury. We created a mouse model with a specific deletion of PAI-1 in the endothelium (PAI-1KOendo) by a Cre-LoxP system. In a model of radiation enteropathy, survival and intestinal radiation injury were followed as well as intestinal gene transcriptional profile and inflammatory cells intestinal infiltration. Irradiated PAI-1KOendo mice exhibited increased survival, reduced acute enteritis severity and attenuated late fibrosis compared with irradiated PAI-1flx/flx mice. Double E-cadherin/TUNEL labeling confirmed a reduced epithelial cell apoptosis in irradiated PAI-1KOendo. High-throughput gene expression combined with bioinformatic analyses revealed a putative involvement of macrophages. We observed a decrease in CD68+cells in irradiated intestinal tissues from PAI-1KOendo mice as well as modifications associated with M1/M2 polarization. This work shows that PAI-1 plays a role in radiation-induced intestinal injury by an endothelium-dependent mechanism and demonstrates in vivo that the endothelium is directly involved in the progression of radiation-induced enteritis.
Collapse
|
137
|
Abe JI, Le NT, Heo KS. Role for SUMOylation in disturbed flow-induced atherosclerotic plaque formation. Biomed Eng Lett 2015. [DOI: 10.1007/s13534-015-0199-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
138
|
Assmann JC, Körbelin J, Schwaninger M. Genetic manipulation of brain endothelial cells in vivo. Biochim Biophys Acta Mol Basis Dis 2015; 1862:381-94. [PMID: 26454206 DOI: 10.1016/j.bbadis.2015.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Julian C Assmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
139
|
Inducible glomerular erythropoietin production in the adult kidney. Kidney Int 2015; 88:1345-1355. [PMID: 26398496 DOI: 10.1038/ki.2015.274] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/26/2015] [Accepted: 07/02/2015] [Indexed: 02/07/2023]
Abstract
Hypoxia-inducible factor (HIF)-2-triggered erythropoietin production in renal interstitial fibroblast-like cells is the physiologically relevant source of erythropoietin for regulating erythropoiesis. During renal fibrosis, these cells transform into myofibroblasts and lose their ability to produce sufficient erythropoietin leading to anemia. To find if other cells for erythropoietin production might exist in the kidney we tested for the capability of nonepithelial glomerular cells to elaborate erythropoietin. Therefore, HIF transcription factors were stabilized by cell-specific deletion of the von Hippel-Lindau (VHL) gene. Inducible deletion of VHL in glomerular connexin40-expressing cells (endothelial, renin-expressing, and mesangial cells) markedly increased glomerular erythropoietin mRNA expression levels, plasma erythropoietin concentrations, and hematocrit values. These changes were mimicked by inducible cell-specific VHL deletion in renin-expressing and in mesangial cells but not in endothelial cells. The increases of erythropoietin production were absent, when VHL was co-deleted with HIF-2. The induction of glomerular erythropoietin expression was associated with the downregulation of juxtaglomerular renin expression, again in a HIF-2-dependent manner. Thus, VHL deletion in renin-expressing and in mesangial cells induces the capability to produce relevant amounts of erythropoietin and to suppress renin expression in the adult kidney if HIF-2 is stabilized.
Collapse
|
140
|
Wang B, Zhao L, Fish M, Logan CY, Nusse R. Self-renewing diploid Axin2(+) cells fuel homeostatic renewal of the liver. Nature 2015; 524:180-5. [PMID: 26245375 PMCID: PMC4589224 DOI: 10.1038/nature14863] [Citation(s) in RCA: 523] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
The source of new hepatocytes in the uninjured liver has remained an open question. By lineage tracing using the Wnt-responsive gene Axin2 in mice, we identify a population of proliferating and self-renewing cells adjacent to the central vein in the liver lobule. These pericentral cells express the early liver progenitor marker Tbx3, are diploid, and thereby differ from mature hepatocytes, which are mostly polyploid. The descendants of pericentral cells differentiate into Tbx3-negative, polyploid hepatocytes, and can replace all hepatocytes along the liver lobule during homeostatic renewal. Adjacent central vein endothelial cells provide Wnt signals that maintain the pericentral cells, thereby constituting the niche. Thus, we identify a cell population in the liver that subserves homeostatic hepatocyte renewal, characterize its anatomical niche, and identify molecular signals that regulate its activity.
Collapse
Affiliation(s)
- Bruce Wang
- 1] Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA [2] Department of Medicine and Liver Center, University of California San Francisco, San Francisco, California 94143, USA
| | - Ludan Zhao
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Matt Fish
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Catriona Y Logan
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Roel Nusse
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
141
|
Chen J, Green J, Yurdagul A, Albert P, McInnis MC, Orr AW. αvβ3 Integrins Mediate Flow-Induced NF-κB Activation, Proinflammatory Gene Expression, and Early Atherogenic Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26212910 DOI: 10.1016/j.ajpath.2015.05.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial cell interactions with transitional matrix proteins, such as fibronectin, occur early during atherogenesis and regulate shear stress-induced endothelial cell activation. Multiple endothelial cell integrins bind transitional matrix proteins, including α5β1, αvβ3, and αvβ5. However, the role these integrins play in mediating shear stress-induced endothelial cell activation remains unclear. Therefore, we sought to elucidate which integrin heterodimers mediate shear stress-induced endothelial cell activation and early atherogenesis. We now show that inhibiting αvβ3 integrins (S247, siRNA), but not α5β1 or αvβ5, blunts shear stress-induced proinflammatory signaling (NF-κB, p21-activated kinase) and gene expression (ICAM1, VCAM1). Importantly, inhibiting αvβ3 did not affect cytokine-induced proinflammatory responses or inhibit all shear stress-induced signaling, because Akt, endothelial nitric oxide synthase, and extracellular regulated kinase activation remained intact. Furthermore, inhibiting αv integrins (S247), but not α5 (ATN-161), in atherosclerosis-prone apolipoprotein E knockout mice significantly reduced vascular remodeling after acute induction of disturbed flow. S247 treatment similarly reduced early diet-induced atherosclerotic plaque formation associated with both diminished inflammation (expression of vascular cell adhesion molecule 1, plaque macrophage content) and reduced smooth muscle incorporation. Inducible, endothelial cell-specific αv integrin deletion similarly blunted inflammation in models of disturbed flow and diet-induced atherogenesis but did not affect smooth muscle incorporation. Our studies identify αvβ3 as the primary integrin heterodimer mediating shear stress-induced proinflammatory responses and as a key contributor to early atherogenic inflammation.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Jonette Green
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Arif Yurdagul
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Patrick Albert
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Marshall C McInnis
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.
| |
Collapse
|
142
|
Delayed inhibition of VEGF signaling after stroke attenuates blood-brain barrier breakdown and improves functional recovery in a comorbidity-dependent manner. J Neurosci 2015; 35:5128-43. [PMID: 25834040 DOI: 10.1523/jneurosci.2810-14.2015] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Diabetes is a common comorbidity in stroke patients and a strong predictor of poor functional outcome. To provide a more mechanistic understanding of this clinically relevant problem, we focused on how diabetes affects blood-brain barrier (BBB) function after stroke. Because the BBB can be compromised for days after stroke and thus further exacerbate ischemic injury, manipulating its function presents a unique opportunity for enhancing stroke recovery long after the window for thrombolytics has passed. Using a mouse model of Type 1 diabetes, we discovered that ischemic stroke leads to an abnormal and persistent increase in vascular endothelial growth factor receptor 2 (VEGF-R2) expression in peri-infarct vascular networks. Correlating with this, BBB permeability was markedly increased in diabetic mice, which could not be prevented with insulin treatment after stroke. Imaging of capillary ultrastructure revealed that BBB permeability was associated with an increase in endothelial transcytosis rather than a loss of tight junctions. Pharmacological inhibition (initiated 2.5 d after stroke) or vascular-specific knockdown of VEGF-R2 after stroke attenuated BBB permeability, loss of synaptic structure in peri-infarct regions, and improved recovery of forepaw function. However, the beneficial effects of VEGF-R2 inhibition on stroke recovery were restricted to diabetic mice and appeared to worsen BBB permeability in nondiabetic mice. Collectively, these results suggest that aberrant VEGF signaling and BBB dysfunction after stroke plays a crucial role in limiting functional recovery in an experimental model of diabetes. Furthermore, our data highlight the need to develop more personalized stroke treatments for a heterogeneous clinical population.
Collapse
|
143
|
Domigan CK, Warren CM, Antanesian V, Happel K, Ziyad S, Lee S, Krall A, Duan L, Torres-Collado AX, Castellani LW, Elashoff D, Christofk HR, van der Bliek AM, Potente M, Iruela-Arispe ML. Autocrine VEGF maintains endothelial survival through regulation of metabolism and autophagy. J Cell Sci 2015; 128:2236-48. [PMID: 25956888 DOI: 10.1242/jcs.163774] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 04/30/2015] [Indexed: 12/12/2022] Open
Abstract
Autocrine VEGF is necessary for endothelial survival, although the cellular mechanisms supporting this function are unknown. Here, we show that--even after full differentiation and maturation--continuous expression of VEGF by endothelial cells is needed to sustain vascular integrity and cellular viability. Depletion of VEGF from the endothelium results in mitochondria fragmentation and suppression of glucose metabolism, leading to increased autophagy that contributes to cell death. Gene-expression profiling showed that endothelial VEGF contributes to the regulation of cell cycle and mitochondrial gene clusters, as well as several--but not all--targets of the transcription factor FOXO1. Indeed, VEGF-deficient endothelium in vitro and in vivo showed increased levels of FOXO1 protein in the nucleus and cytoplasm. Silencing of FOXO1 in VEGF-depleted cells reversed expression profiles of several of the gene clusters that were de-regulated in VEGF knockdown, and rescued both cell death and autophagy phenotypes. Our data suggest that endothelial VEGF maintains vascular homeostasis through regulation of FOXO1 levels, thereby ensuring physiological metabolism and endothelial cell survival.
Collapse
Affiliation(s)
- Courtney K Domigan
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | - Carmen M Warren
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | - Vaspour Antanesian
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | - Katharina Happel
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Safiyyah Ziyad
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | - Sunyoung Lee
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | - Abigail Krall
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Lewei Duan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Antoni X Torres-Collado
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA
| | | | - David Elashoff
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Heather R Christofk
- Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Alexander M van der Bliek
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90024, USA Molecular Biology Institute, University of California, Los Angeles, CA 90024, USA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA
| |
Collapse
|
144
|
Abstract
Coronary artery disease causes acute myocardial infarction and heart failure. Identifying coronary vascular progenitors and their developmental program could inspire novel regenerative treatments for cardiac diseases. The developmental origins of the coronary vessels have been shrouded in mystery and debated for several decades. Recent identification of progenitors for coronary vessels within the endocardium, epicardium, and sinus venosus provides new insights into this question. In addition, significant progress has been achieved in elucidating the cellular and molecular programs that orchestrate coronary artery development. Establishing adequate vascular supply will be an essential component of cardiac regenerative strategies, and these findings raise exciting new strategies for therapeutic cardiac revascularization.
Collapse
Affiliation(s)
- Xueying Tian
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - William T Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
145
|
Integrin β1 controls VE-cadherin localization and blood vessel stability. Nat Commun 2015; 6:6429. [PMID: 25752958 DOI: 10.1038/ncomms7429] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/28/2015] [Indexed: 01/19/2023] Open
Abstract
Angiogenic blood vessel growth requires several distinct but integrated cellular activities. Endothelial cell sprouting and proliferation lead to the expansion of the vasculature and give rise to a highly branched, immature plexus, which is subsequently reorganized into a mature and stable network. Although it is known that integrin-mediated cell-matrix interactions are indispensable for embryonic angiogenesis, little is known about the function of integrins in different steps of vascular morphogenesis. Here, by investigating the integrin β1-subunit with inducible and endothelial-specific gene targeting in the postnatal mouse retina, we show that β1 integrin promotes endothelial sprouting but is a negative regulator of proliferation. In maturing vessels, integrin β1 is indispensable for proper localization of VE-cadherin and thereby cell-cell junction integrity. The sum of our findings establishes that integrin β1 has critical functions in the growing and maturing vasculature, and is required for the formation of stable, non-leaky blood vessels.
Collapse
|
146
|
Whitfield J, Littlewood T, Evan GI, Soucek L. The estrogen receptor fusion system in mouse models: a reversible switch. Cold Spring Harb Protoc 2015; 2015:227-34. [PMID: 25734072 DOI: 10.1101/pdb.top069815] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Reversible regulatory mouse models have significantly contributed to our understanding of normal tissue and cancer biology, providing the opportunity to temporally control initiation, progression, and evolution of physiological and pathological events. The tamoxifen inducible system, one of the best-characterized "reversible switch" models, has a number of beneficial features. In this system, the hormone-binding domain of the mammalian estrogen receptor is used as a heterologous regulatory domain. Upon ligand binding, the receptor is released from its inhibitory complex and the fusion protein becomes functional. We summarize the advantages and drawbacks of the system, describe several mouse models that rely on it, and discuss potential improvements that could render it even more useful and versatile.
Collapse
Affiliation(s)
- Jonathan Whitfield
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain; Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Trevor Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Gerard I Evan
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Laura Soucek
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona 08035, Spain; Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| |
Collapse
|
147
|
Heo KS, Le NT, Cushman HJ, Giancursio CJ, Chang E, Woo CH, Sullivan MA, Taunton J, Yeh ETH, Fujiwara K, Abe JI. Disturbed flow-activated p90RSK kinase accelerates atherosclerosis by inhibiting SENP2 function. J Clin Invest 2015; 125:1299-310. [PMID: 25689261 DOI: 10.1172/jci76453] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 01/06/2015] [Indexed: 01/31/2023] Open
Abstract
Disturbed blood flow (d-flow) causes endothelial cell (EC) dysfunction, leading to atherosclerotic plaque formation. We have previously shown that d-flow increases SUMOylation of p53 and ERK5 through downregulation of sentrin/SUMO-specific protease 2 (SENP2) function; however, it is not known how SENP2 itself is regulated by d-flow. Here, we determined that d-flow activated the serine/threonine kinase p90RSK, which subsequently phosphorylated threonine 368 (T368) of SENP2. T368 phosphorylation promoted nuclear export of SENP2, leading to downregulation of eNOS expression and upregulation of proinflammatory adhesion molecule expression and apoptosis. In an LDLR-deficient murine model of atherosclerosis, EC-specific overexpression of p90RSK increased EC dysfunction and lipid accumulation in the aorta compared with control animals; however, these pathologic changes were not observed in atherosclerotic mice overexpressing dominant negative p90RSK (DN-p90RSK). Moreover, depletion of SENP2 in these mice abolished the protective effect of DN-p90RSK overexpression. We propose that p90RSK-mediated SENP2-T368 phosphorylation is a master switch in d-flow-induced signaling, leading to EC dysfunction and atherosclerosis.
Collapse
|
148
|
Genetic targeting of sprouting angiogenesis using Apln-CreER. Nat Commun 2015; 6:6020. [PMID: 25597280 PMCID: PMC4309445 DOI: 10.1038/ncomms7020] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/02/2014] [Indexed: 02/07/2023] Open
Abstract
Under pathophysiological conditions in adults, endothelial cells (ECs) sprout from pre-existing blood vessels to form new ones by a process termed angiogenesis. During embryonic development, Apelin (APLN) is robustly expressed in vascular ECs. In adult mice, however, APLN expression in the vasculature is significantly reduced. Here we show that APLN expression is reactivated in adult ECs after ischaemia insults. In models of both injury ischaemia and tumor angiogenesis, we find that Apln-CreER genetically labels sprouting but not quiescent vasculature. By leveraging this specific activity, we demonstrate that abolishment of the VEGF-VEGFR2 signalling pathway as well as ablation of sprouting ECs diminished tumour vascularization and growth without compromising vascular homeostasis in other organs. Collectively, we show that Apln-CreER distinguishes sprouting vessels from stabilized vessels in multiple pathological settings. The Apln-CreER line described here will greatly aid future mechanistic studies in both vascular developmental biology and adult vascular diseases.
Collapse
|
149
|
The Wilms' tumour suppressor Wt1 is a major regulator of tumour angiogenesis and progression. Nat Commun 2014; 5:5852. [PMID: 25510679 DOI: 10.1038/ncomms6852] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, activation of metastasis and avoidance of immune destruction are important for cancer progression. These biological capabilities are, apart from cancer cells, mediated by different cell types, including endothelial, haematopoietic progenitor and myeloid-derived suppressor cells. We show here that all these cell types frequently express the Wilms' tumour suppressor Wt1, which transcriptionally controls expression of Pecam-1 (CD31) and c-kit (CD117). Inducible conditional knockout of Wt1 in endothelial, haematopoietic and myeloid-derived suppressor cells is sufficient to cause regression of tumour vascularization and an enhanced immune response, leading to decreased metastasis, regression of established tumours and enhanced survival. Thus, Wt1 is an important regulator of cancer growth via modulation of tumour vascularization, immune response and metastasis formation.
Collapse
|
150
|
Han Y, Tao J, Gomer A, Ramirez-Bergeron DL. Loss of endothelial-ARNT in adult mice contributes to dampened circulating proangiogenic cells and delayed wound healing. Vasc Med 2014; 19:429-41. [PMID: 25398385 DOI: 10.1177/1358863x14559588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The recruitment and homing of circulating bone marrow-derived cells include endothelial progenitor cells (EPCs) that are critical to neovascularization and tissue regeneration of various vascular pathologies. We report here that conditional inactivation of hypoxia-inducible factor's (HIF) transcriptional activity in the endothelium of adult mice (Arnt(ΔiEC) mice) results in a disturbance of infiltrating cells, a hallmark of neoangiogenesis, during the early phases of wound healing. Cutaneous biopsy punches show distinct migration of CD31(+) cells into wounds of control mice by 36 hours. However, a significant decline in numbers of infiltrating cells with immature vascular markers, as well as decreased transcript levels of genes associated with their expression and recruitment, were identified in wounds of Arnt(ΔiEC) mice. Matrigel plug assays further confirmed neoangiogenic deficiencies alongside a reduction in numbers of proangiogenic progenitor cells from bone marrow and peripheral blood samples of recombinant vascular endothelial growth factor-treated Arnt(ΔiEC) mice. In addition to HIF's autocrine requirements in endothelial cells, our data implicate that extrinsic microenvironmental cues provided by endothelial HIF are pivotal for early migration of proangiogenic cells, including those involved in wound healing.
Collapse
Affiliation(s)
- Yu Han
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Jiayi Tao
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alla Gomer
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Diana L Ramirez-Bergeron
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|