1
|
Keogh K, Coen S, Lonergan P, Fair S, Kenny DA. Complement 3 (C3) within the hypothalamic arcuate nucleus is a potential key mediator of the effect of enhanced nutrition on reproductive development in young bull calves. BMC Genomics 2025; 26:466. [PMID: 40346477 PMCID: PMC12065335 DOI: 10.1186/s12864-025-11656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Reproductive development may be advanced in bull calves through enhanced dietary intake during the early life period. This effect between enhanced nutrition with subsequent earlier reproductive development is orchestrated through signalling within the hypothalamic-pituitary-testicular axis. Within the hypothalamus, the arcuate nucleus (ARC) is crucial for the integration of peripheral metabolic status with subsequent gonadotropin releasing hormone (GnRH) signalling; however, the precise molecular control regulating this effect is not fully known. The aim of this study was to evaluate the global transcriptomic and proteomic responses to varied plane of nutrition during early calf-hood in young dairy bull calves. Additionally, we sought to integrate these 'omics' datasets to determine key genes and proteins contributing to earlier reproductive development. Between 2-12 weeks of age, 30 Holstein-Friesian bull calves (mean age: 17.5 days; mean bodyweight 48.8 kg), were offered either a high or moderate plane of nutrition with 15 calves in each group. At 12 weeks of age, all calves were euthanised and the ARC tissue isolated from each calf. The ARC tissue was then used for global transcriptomic (miRNAseq and mRNAseq) and proteomic analyses. RESULTS Bioinformatic analyses were undertaken to determine differentially expressed transcripts (FDR < 0.1; fold change > 1.5) between the dietary treatment groups, resulting in the identification of 1 differentially expressed miRNA (miR-2419-3p) and 83 differentially expressed mRNA in the ARC region. mRNA target gene prediction identified Complement 3 (C3) as a target of miR-2419-3p, suggesting a relationship between the two transcripts. Furthermore, through a co-regulatory network analysis conducted on the proteomics dataset, C3 was revealed as a hub protein. Additionally, through the proteomic network analysis, C3 was interacting with proteins involved in both insulin and GnRH signalling, highlighting a potential role for C3 in mediated the effect of enhanced nutritional status with earlier reproductive development within the ARC. CONCLUSION This study highlights an effect of altered plane of nutrition in early life on the molecular control of the hypothalamic ARC. Additionally, results generated suggest a potential role for the C3 gene in mediating the interaction between enhanced metabolic status with reproductive development within the ARC, regulated by miR-2419-3p.
Collapse
Affiliation(s)
- Kate Keogh
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
| | - Stephen Coen
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean Fair
- Laboratory of Animal Reproduction, Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - David A Kenny
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland.
| |
Collapse
|
2
|
Aksan B, Mauceri D. Beyond vessels: unraveling the impact of VEGFs on neuronal functions and structure. J Biomed Sci 2025; 32:33. [PMID: 40050849 PMCID: PMC11884128 DOI: 10.1186/s12929-025-01128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
Neurons rely on the bloodstream for essential nutrients and oxygen, which is facilitated by an intricate coupling of the neuronal and vascular systems. Central to this neurovascular interaction is the vascular endothelial growth factor (VEGF) family, a group of secreted growth factors traditionally known for their roles in promoting endothelial cell proliferation, migration, and survival in the cardiovascular and lymphatic systems. However, emerging evidence shows that VEGFs also play indispensable roles in the nervous system, extending beyond their canonical angiogenic and lymphangiogenic functions. Over the past two decades, VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells. In particular, it has become increasingly evident that VEGFs also play crucial functions in the development, regulation, and maintenance of neuronal morphology. Understanding the roles of VEGFs in neuronal development is of high scientific and clinical interest because of the significance of precise neuronal morphology for neural connectivity and network function, as well as the association of morphological abnormalities with neurological and neurodegenerative disorders. This review begins with an overview of the VEGF family members, their structural characteristics, receptors, and established roles in vasculature. However, it then highlights and focuses on the exciting variety of neuronal functions of VEGFs, especially their crucial role in the development, regulation, and maintenance of neuronal morphology.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Institute of Anatomy and Cell Biology, Dept. Molecular and Cellular Neuroscience, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
3
|
Smith GT, Radin DP, Tsirka SE. From protein-protein interactions to immune modulation: Therapeutic prospects of targeting Neuropilin-1 in high-grade glioma. Front Immunol 2022; 13:958620. [PMID: 36203599 PMCID: PMC9532003 DOI: 10.3389/fimmu.2022.958620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the past several years there has been a marked increase in our understanding of the pathophysiological hallmarks of glioblastoma development and progression, with specific respect to the contribution of the glioma tumor microenvironment to the rapid progression and treatment resistance of high-grade gliomas. Despite these strides, standard of care therapy still only targets rapidly dividing tumor cells in the glioma, and does little to curb the pro-tumorigenic functions of non-cancerous cells entrenched in the glioma microenvironment. This tumor promoting environment as well as the heterogeneity of high-grade gliomas contribute to the poor prognosis of this malignancy. The interaction of non-malignant cells in the microenvironment with the tumor cells accentuate phenotypes such as rapid proliferation or immunosuppression, so therapeutically modulating one target expressed on one cell type may be insufficient to restrain these rapidly developing neoplasias. With this in mind, identifying a target expressed on multiple cell types and understanding how it governs tumor-promoting functions in each cell type may have great utility in better managing this disease. Herein, we review the physiology and pathological effects of Neuropilin-1, a transmembrane co-receptor which mediates signal transduction pathways when associated with multiple other receptors. We discuss its effects on the properties of endothelial cells and on immune cell types within gliomas including glioma-associated macrophages, microglia, cytotoxic T cells and T regulatory cells. We also consider its effects when elaborated on the surface of tumor cells with respect to proliferation, stemness and treatment resistance, and review attempts to target Neuroplin-1 in the clinical setting.
Collapse
Affiliation(s)
- Gregory T. Smith
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Daniel P. Radin
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Stella E. Tsirka
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- *Correspondence: Stella E. Tsirka,
| |
Collapse
|
4
|
Duittoz AH, Tillet Y, Geller S. The great migration: how glial cells could regulate GnRH neuron development and shape adult reproductive life. J Chem Neuroanat 2022; 125:102149. [PMID: 36058434 DOI: 10.1016/j.jchemneu.2022.102149] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 10/31/2022]
Abstract
In mammals, reproductive function is under the control of hypothalamic neurons named Gonadotropin-Releasing Hormone (GnRH) neurons. These neurons migrate from the olfactory placode to the brain, during embryonic development. For the past 40 years, these neurons have been considered an example of tangential migration, i.e., dependent on the olfactory/vomeronasal/terminal nerves. Numerous studies have highlighted the factors involved in the migration of these neurons but thus far overlooked the cellular microenvironment that produces them. Many of these factors are dysregulated in hypogonadotropic hypogonadism, resulting in subfertility/infertility. Nevertheless, over the past ten years, several papers have reported the influence of glial cells (named olfactory ensheathing cells [OECs]) in the migration and differentiation of GnRH neurons. This review will describe the atypical origins, migration, and differentiation of these neurons, focusing on the latest discoveries. There will be a more specific discussion on the involvement of OECs in the development of GnRH neurons, during embryonic and perinatal life; as well as on their potential implication in the development of congenital or idiopathic hypogonadotropic hypogonadism (such as Kallmann syndrome).
Collapse
Affiliation(s)
- Anne H Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Yves Tillet
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRA Val de Loire, Université de Tours, IFCE, 37380 Nouzilly, France
| | - Sarah Geller
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
5
|
Yellapragada V, Eskici N, Wang Y, Madhusudan S, Vaaralahti K, Tuuri T, Raivio T. Time and dose-dependent effects of FGF8-FGFR1 signaling in GnRH neurons derived from human pluripotent stem cells. Dis Model Mech 2022; 15:276003. [PMID: 35833364 PMCID: PMC9403748 DOI: 10.1242/dmm.049436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper. Summary: This article demonstrates the essential role FGF8–FGFR1 signaling has in the development of gonadotropin-releasing hormone (GnRH)-expressing neurons by using a human stem cell model.
Collapse
Affiliation(s)
- Venkatram Yellapragada
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Yafei Wang
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, 00029 Helsinki University Hospital, Helsinki, Finland
| | - Taneli Raivio
- Stem Cells and Metabolism Research Program (STEMM), Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,Medicum, Faculty of Medicine, 00014 University of Helsinki, Helsinki, Finland.,New Children's Hospital, Pediatric Research Center, 00029 Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
6
|
Delayed Therapeutic Administration of Melatonin Enhances Neuronal Survival Through AKT and MAPK Signaling Pathways Following Focal Brain Ischemia in Mice. J Mol Neurosci 2022; 72:994-1007. [PMID: 35307786 DOI: 10.1007/s12031-022-01995-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 03/01/2022] [Indexed: 10/18/2022]
Abstract
Melatonin has a role in the cell survival signaling pathways as a candidate for secondary stroke prevention. Therefore, in the present study, the coordination of ipsilateral and contralateral hemispheres to evaluate delayed post-acute effect of melatonin was examined on recovery of the cell survival and apoptosis after stroke. Melatonin was administered (4 mg/kg/day) intraperitoneally for 45 days, starting 3 days after 30 min of middle cerebral artery occlusion. The genes and proteins related to the cell survival and apoptosis were investigated by immunofluorescence, western blotting, and RT-PCR techniques after behavioral experiments. Melatonin produced delayed neurological recovery by improving motor coordination on grip strength and rotarod tests. This neurological recovery was also reflected by high level of NeuN positive cells and low level of TUNEL-positive cells suggesting enhanced neuronal survival and reduced apoptosis at the fifty-fifth day of stroke. The increase of NGF, Nrp1, c-jun; activation of AKT; and dephosphorylation of ERK and JNK at the fifty-fifth day showed that cell survival and apoptosis signaling molecules compete to contribute to the remodeling of brain. Furthermore, an increase in the CREB and Atf-1 expressions suggested the melatonin's strong reformative effect on neuronal regeneration. The contralateral hemisphere was more active at the latter stages of the molecular and functional regeneration which provides a further proof of principle about melatonin's action on the promotion of brain plasticity and recovery after stroke.
Collapse
|
7
|
Oda A, Inoue S, Kaneko R, Narita Y, Shiono S, Kaneko T, Tseng YC, Ohtani-Kaneko R. Involvement of IGF-1R-PI3K-AKT-mTOR pathway in increased number of GnRH3 neurons during androgen-induced sex reversal of the brain in female tilapia. Sci Rep 2022; 12:2450. [PMID: 35165334 PMCID: PMC8844422 DOI: 10.1038/s41598-022-06384-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
The neuroplastic mechanism of sex reversal in the fish brain remains unclear due to the difficulty in identifying the key neurons involved. Mozambique tilapia show different reproductive behaviours between sexes; males build circular breeding nests while females hold and brood fertilized eggs in their mouth. In tilapia, gonadotropin-releasing hormone 3 (GnRH3) neurons, located in the terminal nerve, regulate male reproductive behaviour. Mature males have more GnRH3 neurons than mature females, and these neurons have been indicated to play a key role in the androgen-induced female-to-male sex reversal of the brain. We aimed to elucidate the signalling pathway involved in the androgen-induced increase in GnRH3 neurons in mature female tilapia. Applying inhibitors to organotypic cultures of brain slices, we showed that the insulin-like growth factor (IGF)-1 receptor (IGF-1R)/PI3K/AKT/mTOR pathway contributed to the androgen-induced increase in GnRH3 neurons. The involvement of IGF-1 and IGF-1R in 11-ketotestosterone (11-KT)-induced development of GnRH3 neurons was supported by an increase in Igf-1 mRNA shortly after 11-KT treatment, the increase of GnRH3 neurons after IGF-1 treatment and the expression of IGF-1R in GnRH3 neurons. Our findings highlight the involvement of IGF-1 and its downstream signalling pathway in the sex reversal of the tilapia brain.
Collapse
|
8
|
Saleki K, Banazadeh M, Miri NS, Azadmehr A. Triangle of cytokine storm, central nervous system involvement, and viral infection in COVID-19: the role of sFasL and neuropilin-1. Rev Neurosci 2021; 33:147-160. [PMID: 34225390 DOI: 10.1515/revneuro-2021-0047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) is identified as the cause of coronavirus disease 2019 (COVID-19), and is often linked to extreme inflammatory responses by over activation of neutrophil extracellular traps (NETs), cytokine storm, and sepsis. These are robust causes for multi-organ damage. In particular, potential routes of SARS-CoV2 entry, such as angiotensin-converting enzyme 2 (ACE2), have been linked to central nervous system (CNS) involvement. CNS has been recognized as one of the most susceptible compartments to cytokine storm, which can be affected by neuropilin-1 (NRP-1). ACE2 is widely-recognized as a SARS-CoV2 entry pathway; However, NRP-1 has been recently introduced as a novel path of viral entry. Apoptosis of cells invaded by this virus involves Fas receptor-Fas ligand (FasL) signaling; moreover, Fas receptor may function as a controller of inflammation. Furthermore, NRP-1 may influence FasL and modulate cytokine profile. The neuroimmunological insult by SARS-CoV2 infection may be inhibited by therapeutic approaches targeting soluble Fas ligand (sFasL), cytokine storm elements, or related viral entry pathways. In the current review, we explain pivotal players behind the activation of cytokine storm that are associated with vast CNS injury. We also hypothesize that sFasL may affect neuroinflammatory processes and trigger the cytokine storm in COVID-19.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, 76169-13555, Iran
| | - Niloufar Sadat Miri
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| | - Abbas Azadmehr
- National Elite Foundation, Mazandaran Province Branch, Tehran, 48157-66435, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, 47176-47745, Iran
- Medical Immunology Department, Babol University of Medical Sciences, Babol, 47176-47745, Iran
| |
Collapse
|
9
|
Onesto MM, Short CA, Rempel SK, Catlett TS, Gomez TM. Growth Factors as Axon Guidance Molecules: Lessons From in vitro Studies. Front Neurosci 2021; 15:678454. [PMID: 34093120 PMCID: PMC8175860 DOI: 10.3389/fnins.2021.678454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Growth cones at the tips of extending axons navigate through developing organisms by probing extracellular cues, which guide them through intermediate steps and onto final synaptic target sites. Widespread focus on a few guidance cue families has historically overshadowed potentially crucial roles of less well-studied growth factors in axon guidance. In fact, recent evidence suggests that a variety of growth factors have the ability to guide axons, affecting the targeting and morphogenesis of growth cones in vitro. This review summarizes in vitro experiments identifying responses and signaling mechanisms underlying axon morphogenesis caused by underappreciated growth factors.
Collapse
Affiliation(s)
| | | | | | | | - Timothy M. Gomez
- Neuroscience Training Program and Cell and Molecular Biology Program, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
10
|
Abstract
Many of the recent advances in our understanding of human reproductive biology and its genetic basis have arisen directly via the genetic investigation of patients with Kallmann syndrome and their families. The disease is characterised by the association of an isolated defect in the secretion (or, less commonly, action) of gonadotropin-releasing hormone (GnRH) and consequent infertility, with anosmia and potentially other associated non-reproductive features. GnRH-producing neurons are located in the hypothalamic brain region after a peculiar migration during embryonic life. To date, different genes affecting GnRH neuron development/migration have so far been implicated in Kallmann syndrome, but our knowledge of the genetic basis of the syndrome remains incomplete. From a clinical point of view, the disease has suffered from a lack of definitive diagnosis and treatment, and although progress has been made in terms of timely diagnosis and evidence-based treatment of patients, implementation remains inconsistent. These aspects will be discussed in this review, which examines new strategies for arriving at more evidence-based and patient-centred medical practice in Kallmann syndrome.
Collapse
Affiliation(s)
- Du Soon Swee
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Richard Quinton
- Department of Endocrinology, Diabetes & Metabolism, Royal Victoria Infirmary, Newcastle-Upon-Tyne Hospitals, Newcastle-upon-Tyne, UK
- Translational & Clinical Research Institute, University of Newcastle-upon-Tyne, Newcastle-Upon-Tyne, UK
| | - Roberto Maggi
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, MI, Italy
| |
Collapse
|
11
|
Short CA, Onesto MM, Rempel SK, Catlett TS, Gomez TM. Familiar growth factors have diverse roles in neural network assembly. Curr Opin Neurobiol 2021; 66:233-239. [PMID: 33477094 PMCID: PMC8058242 DOI: 10.1016/j.conb.2020.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
The assembly of neuronal circuits during development depends on guidance of axonal growth cones by molecular cues deposited in their environment. While a number of families of axon guidance molecules have been identified and reviewed, important and diverse activities of traditional growth factors are emerging. Besides clear and well recognized roles in the regulation of cell division, differentiation and survival, new research shows later phase roles for a number of growth factors in promoting neuronal migration, axon guidance and synapse formation throughout the nervous system.
Collapse
Affiliation(s)
- Caitlin A Short
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Massimo M Onesto
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States; Stanford University School of Medicine, United States
| | - Sarah K Rempel
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Timothy S Catlett
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States
| | - Timothy M Gomez
- Department of Neuroscience and Neuroscience Training Program, University of Wisconsin, Madison, WI 53706, United States.
| |
Collapse
|
12
|
Abstract
Neuropilin-1 (NRP-1), a member of a family of signaling proteins, was shown to serve as an entry factor and potentiate SARS Coronavirus 2 (SARS-CoV-2) infectivity in vitro. This cell surface receptor with its disseminated expression is important in angiogenesis, tumor progression, viral entry, axonal guidance, and immune function. NRP-1 is implicated in several aspects of a SARS-CoV-2 infection including possible spread through the olfactory bulb and into the central nervous system and increased NRP-1 RNA expression in lungs of severe Coronavirus Disease 2019 (COVID-19). Up-regulation of NRP-1 protein in diabetic kidney cells hint at its importance in a population at risk of severe COVID-19. Involvement of NRP-1 in immune function is compelling, given the role of an exaggerated immune response in disease severity and deaths due to COVID-19. NRP-1 has been suggested to be an immune checkpoint of T cell memory. It is unknown whether involvement and up-regulation of NRP-1 in COVID-19 may translate into disease outcome and long-term consequences, including possible immune dysfunction. It is prudent to further research NRP-1 and its possibility of serving as a therapeutic target in SARS-CoV-2 infections. We anticipate that widespread expression, abundance in the respiratory and olfactory epithelium, and the functionalities of NRP-1 factor into the multiple systemic effects of COVID-19 and challenges we face in management of disease and potential long-term sequelae.
Collapse
Affiliation(s)
- Bindu S. Mayi
- Department of Basic Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, Florida, United States of America
- * E-mail:
| | - Jillian A. Leibowitz
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Arden T. Woods
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Katherine A. Ammon
- USF Morsani College of Medicine, Tampa, Florida, United States of America
| | - Alphonse E. Liu
- Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, Florida, United States of America
| | - Aarti Raja
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| |
Collapse
|
13
|
Oleari R, André V, Lettieri A, Tahir S, Roth L, Paganoni A, Eberini I, Parravicini C, Scagliotti V, Cotellessa L, Bedogni F, De Martini LB, Corridori MV, Gulli S, Augustin HG, Gaston-Massuet C, Hussain K, Cariboni A. A Novel SEMA3G Mutation in Two Siblings Affected by Syndromic GnRH Deficiency. Neuroendocrinology 2021; 111:421-441. [PMID: 32365351 DOI: 10.1159/000508375] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 05/01/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Gonadotropin-releasing hormone (GnRH) deficiency causes hypogonadotropic hypogonadism (HH), a rare genetic disorder that impairs sexual reproduction. HH can be due to defective GnRH-secreting neuron development or function and may be associated with other clinical signs in overlapping genetic syndromes. With most of the cases being idiopathic, genetics underlying HH is still largely unknown. OBJECTIVE To assess the contribution of mutated Semaphorin 3G (SEMA3G) in the onset of a syndromic form of HH, characterized by intellectual disability and facial dysmorphic features. METHOD By combining homozygosity mapping with exome sequencing, we identified a novel variant in the SEMA3G gene. We then applied mouse as a model organism to examine SEMA3Gexpression and its functional requirement in vivo. Further, we applied homology modelling in silico and cell culture assays in vitro to validate the pathogenicity of the identified gene variant. RESULTS We found that (i) SEMA3G is expressed along the migratory route of GnRH neurons and in the developing pituitary, (ii) SEMA3G affects GnRH neuron development, but is redundant in the adult hypothalamic-pituitary-gonadal axis, and (iii) mutated SEMA3G alters binding properties in silico and in vitro to its PlexinA receptors and attenuates its effect on the migration of immortalized GnRH neurons. CONCLUSION In silico, in vitro, and in vivo models revealed that SEMA3G regulates GnRH neuron migration and that its mutation affecting receptor selectivity may be responsible for the HH-related defects.
Collapse
Affiliation(s)
- Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Valentina André
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Sophia Tahir
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Lise Roth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Alyssa Paganoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Chiara Parravicini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Valeria Scagliotti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ludovica Cotellessa
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Laboratory of Endocrine and Metabolic Research, Milan, Italy
| | - Francesco Bedogni
- San Raffaele Rett Research Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff, United Kingdom
| | | | | | - Simona Gulli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Khalid Hussain
- Sidra Medical & Research Center, Division of Endocrinology OPC, Department of Pediatric Medicine, Doha, Qatar
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy,
| |
Collapse
|
14
|
Gbx2 Is Required for the Migration and Survival of a Subpopulation of Trigeminal Cranial Neural Crest Cells. J Dev Biol 2020; 8:jdb8040033. [PMID: 33322598 PMCID: PMC7768483 DOI: 10.3390/jdb8040033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/30/2022] Open
Abstract
The development of key structures within the mature vertebrate hindbrain requires the migration of neural crest (NC) cells and motor neurons to their appropriate target sites. Functional analyses in multiple species have revealed a requirement for the transcription factor gastrulation-brain-homeobox 2 (Gbx2) in NC cell migration and positioning of motor neurons in the developing hindbrain. In addition, loss of Gbx2 function studies in mutant mouse embryos, Gbx2neo, demonstrate a requirement for Gbx2 for the development of NC-derived sensory neurons and axons constituting the mandibular branch of the trigeminal nerve (CNV). Our recent GBX2 target gene identification study identified multiple genes required for the migration and survival of NC cells (e.g., Robo1, Slit3, Nrp1). In this report, we performed loss-of-function analyses using Gbx2neo mutant embryos, to improve our understanding of the molecular and genetic mechanisms regulated by Gbx2 during anterior hindbrain and CNV development. Analysis of Tbx20 expression in the hindbrain of Gbx2neo homozygotes revealed a severely truncated rhombomere (r)2. Our data also provide evidence demonstrating a requirement for Gbx2 in the temporal regulation of Krox20 expression in r3. Lastly, we show that Gbx2 is required for the expression of Nrp1 in a subpopulation of trigeminal NC cells, and correct migration and survival of cranial NC cells that populate the trigeminal ganglion. Taken together, these findings provide additional insight into molecular and genetic mechanisms regulated by Gbx2 that underlie NC migration, trigeminal ganglion assembly, and, more broadly, anterior hindbrain development.
Collapse
|
15
|
Yoshino Y, Roy B, Dwivedi Y. Altered miRNA landscape of the anterior cingulate cortex is associated with potential loss of key neuronal functions in depressed brain. Eur Neuropsychopharmacol 2020; 40:70-84. [PMID: 32600964 PMCID: PMC7655604 DOI: 10.1016/j.euroneuro.2020.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/05/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs), a family of non-coding RNAs, have recently gained a considerable attention in neuropsychiatric disorders. Being a pleiotropic modulator of target gene(s), miRNA has been recognized as central to downstream gene regulatory networks. In the recent past, reports have suggested their role in changing the epigenetic landscape in brain of subjects with major depressive disorder (MDD). Anterior cingulate cortex (ACC) is a brain area implicated in several complex cognitive functions, such as impulse control, emotion, and decision-making and is associated with psychopathology associated with mood regulation. In this study, we examined whether MDD is associated with altered miRNA transcriptome in ACC and whether altered miRNA landscape is associated with modifications in specific gene network(s) at the functional level. Using next generation sequencing (NGS), it was observed that 117 miRNAs (4.61%) were significantly upregulated and 54 (2.13%) were downregulated in MDD subjects (n = 22) compared with non-psychiatric controls (n = 25). Using 24 most significantly upregulated miRNAs in the MDD group, we determined functional enrichment of target genes and found them to be associated with long-term potentiation, neurotrophin signaling, and axon guidance. Intra- and inter-cluster similarities of enriched terms based on overrepresented gene list showed neurobiological functions associated with neuronal growth and survival. Web centric parameters and ontology enrichment functions identified two major domains related to phosphatidyl signaling, GTPase signaling, neuronal migration, and neurotrophin signaling. Our findings of altered miRNA landscape along with a shift in targetome relate to previously reported morphometric changes and neuronal atrophy in ACC of MDD subjects.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
16
|
Vanacker C, Trova S, Shruti S, Casoni F, Messina A, Croizier S, Malone S, Ternier G, Hanchate NK, Rasika S, Bouret SG, Ciofi P, Giacobini P, Prevot V. Neuropilin-1 expression in GnRH neurons regulates prepubertal weight gain and sexual attraction. EMBO J 2020; 39:e104633. [PMID: 32761635 PMCID: PMC7527814 DOI: 10.15252/embj.2020104633] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022] Open
Abstract
Hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH), the "master molecule" regulating reproduction and fertility, migrate from their birthplace in the nose to their destination using a system of guidance cues, which include the semaphorins and their receptors, the neuropilins and plexins, among others. Here, we show that selectively deleting neuropilin-1 in new GnRH neurons enhances their survival and migration, resulting in excess neurons in the hypothalamus and in their unusual accumulation in the accessory olfactory bulb, as well as an acceleration of mature patterns of activity. In female mice, these alterations result in early prepubertal weight gain, premature attraction to male odors, and precocious puberty. Our findings suggest that rather than being influenced by peripheral energy state, GnRH neurons themselves, through neuropilin-semaphorin signaling, might engineer the timing of puberty by regulating peripheral adiposity and behavioral switches, thus acting as a bridge between the reproductive and metabolic axes.
Collapse
Affiliation(s)
- Charlotte Vanacker
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sonal Shruti
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Filippo Casoni
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Andrea Messina
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sophie Croizier
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Samuel Malone
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Naresh Kumar Hanchate
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Sebastien G Bouret
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Philippe Ciofi
- Inserm U1215Neurocentre MagendieBordeauxFrance
- Université de BordeauxBordeauxFrance
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine BrainUniv. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR‐S 1172LilleFrance
- FHU, 1000 Days for HealthLilleFrance
| |
Collapse
|
17
|
Kay VR, Rätsep MT, Figueiró-Filho EA, Croy BA. Preeclampsia may influence offspring neuroanatomy and cognitive function: a role for placental growth factor†. Biol Reprod 2020; 101:271-283. [PMID: 31175349 DOI: 10.1093/biolre/ioz095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Abstract
Preeclampsia (PE) is a common pregnancy complication affecting 3-5% of women. Preeclampsia is diagnosed clinically as new-onset hypertension with associated end organ damage after 20 weeks of gestation. Despite being diagnosed as a maternal syndrome, fetal experience of PE is a developmental insult with lifelong cognitive consequences. These cognitive alterations are associated with distorted neuroanatomy and cerebrovasculature, including a higher risk of stroke. The pathophysiology of a PE pregnancy is complex, with many factors potentially able to affect fetal development. Deficient pro-angiogenic factor expression is one aspect that may impair fetal vascularization, alter brain structure, and affect future cognition. Of the pro-angiogenic growth factors, placental growth factor (PGF) is strongly linked to PE. Concentrations of PGF are inappropriately low in maternal blood both before and during a PE gestation. Fetal concentrations of PGF appear to mirror maternal circulating concentrations. Using Pgf-/- mice that may model effects of PE on offspring, we demonstrated altered central nervous system vascularization, neuroanatomy, and behavior. Overall, we propose that development of the fetal brain is impaired in PE, making the offspring of preeclamptic pregnancies a unique cohort with greater risk of altered cognition and cerebrovasculature. These individuals may benefit from early interventions, either pharmacological or environmental. The early neonatal period may be a promising window for intervention while the developing brain retains plasticity.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Matthew T Rätsep
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
18
|
Balashova L, Bykovskaya S, Korobova L, Kuznetsova Y, Kantardgy E, Mukhin V, Popov A. Immunological outcomes in infants with ROP after dexamethasone and aminophylline. Clin Exp Pharmacol Physiol 2020; 47:1368-1373. [PMID: 32198937 DOI: 10.1111/1440-1681.13308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/18/2020] [Accepted: 03/18/2020] [Indexed: 11/28/2022]
Abstract
This research aims to improve anaesthesia services given to preterm infants by the use of dexamethasone and aminophylline administrated under sevoflurane, and to analyze its effect on the cell-mediated immunity (CD4+CD25+Foxp3+(T-reg) and CD4+CD25highFoxp3+CD127low). We have examined 74 premature babies with retinopathy of prematurity (ROP) at the 3-5 stages during the 25-32 week gestation period (1-6 months after birth). Both immunomodulators had no significant effect on clinical parameters after one dose (P > .05). Aminophylline (2.4% solution, 0.1 mL/kg or 0.132 mL per infant on average) and dexamethasone (0.4% solution, 0.1 mg/kg or 0.132 mL per infant on average) were intravenously injected 15 minutes before the end of the surgery. Required anaesthesia depth was maintained with inhalation anaesthetic (1.5-2.0 IAC), and the minimum fresh gas flow was not less than 2 L. Blood samples were taken from the vein (anaesthesia induction stage) into the tubes containing EDTA (the anticoagulant), stored at 20-25°C, and then, processed and stained within 24 hours after sampling. Both immunomodulators had no significant effect on clinical parameters after one dose (P > .05). Short-term shift in regulatory T-cell level affected by dexamethasone has a negative effect combined with further withdrawal effect that this hormonal drug has. Aminophylline has such clinical effects as improving pulmonary ventilation, decrease in apnoea frequency, and improving blood gas indices. Aminophylline has less expressed but more prolonged positive effect during the day when used for several days. It may lead to a persistent positive effect with progressive treatment outcomes.
Collapse
Affiliation(s)
- Larisa Balashova
- Library of Children's Ophthalmology, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Svetlana Bykovskaya
- Departament of Cell's Technology and Regenerative Medicine, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Ludmila Korobova
- Departament of Anesthesiology, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Yulia Kuznetsova
- Department of Ophthalmology, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Elena Kantardgy
- Library of Children's Ophthalmology, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Vladimir Mukhin
- Departament of Cell's Technology and Regenerative Medicine, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| | - Andrey Popov
- Library of Children's Ophthalmology, Pirogov Russian National Research Medical University (PRNIMU), Moscow, Russia
| |
Collapse
|
19
|
Mancini A, Howard SR, Marelli F, Cabrera CP, Barnes MR, Sternberg MJ, Leprovots M, Hadjidemetriou I, Monti E, David A, Wehkalampi K, Oleari R, Lettieri A, Vezzoli V, Vassart G, Cariboni A, Bonomi M, Garcia MI, Guasti L, Dunkel L. LGR4 deficiency results in delayed puberty through impaired Wnt/β-catenin signaling. JCI Insight 2020; 5:133434. [PMID: 32493844 PMCID: PMC7308048 DOI: 10.1172/jci.insight.133434] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
The initiation of puberty is driven by an upsurge in hypothalamic gonadotropin-releasing hormone (GnRH) secretion. In turn, GnRH secretion upsurge depends on the development of a complex GnRH neuroendocrine network during embryonic life. Although delayed puberty (DP) affects up to 2% of the population, is highly heritable, and is associated with adverse health outcomes, the genes underlying DP remain largely unknown. We aimed to discover regulators by whole-exome sequencing of 160 individuals of 67 multigenerational families in our large, accurately phenotyped DP cohort. LGR4 was the only gene remaining after analysis that was significantly enriched for potentially pathogenic, rare variants in 6 probands. Expression analysis identified specific Lgr4 expression at the site of GnRH neuron development. LGR4 mutant proteins showed impaired Wnt/β-catenin signaling, owing to defective protein expression, trafficking, and degradation. Mice deficient in Lgr4 had significantly delayed onset of puberty and fewer GnRH neurons compared with WT, whereas lgr4 knockdown in zebrafish embryos prevented formation and migration of GnRH neurons. Further, genetic lineage tracing showed strong Lgr4-mediated Wnt/β-catenin signaling pathway activation during GnRH neuron development. In conclusion, our results show that LGR4 deficiency impairs Wnt/β-catenin signaling with observed defects in GnRH neuron development, resulting in a DP phenotype. Defects of LGR4/Wnt-β-catenin activity compromise the development of the GnRH neuroendocrine network, resulting in delayed onset of puberty in humans and mice.
Collapse
Affiliation(s)
- Alessandra Mancini
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Federica Marelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | - Claudia P Cabrera
- Centre for Translational Bioinformatics, William Harvey Research Institute, and.,NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michael R Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, and.,NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michael Je Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elena Monti
- St George's NHS Foundation Trust, London, United Kingdom
| | - Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Karoliina Wehkalampi
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Valeria Vezzoli
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | | | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Marco Bonomi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Milan, Italy
| | | | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
20
|
Grandi FC, De Tomasi L, Mustapha M. Single-Cell RNA Analysis of Type I Spiral Ganglion Neurons Reveals a Lmx1a Population in the Cochlea. Front Mol Neurosci 2020; 13:83. [PMID: 32523514 PMCID: PMC7261882 DOI: 10.3389/fnmol.2020.00083] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
In the mature cochlea, each inner hair cell (IHC) is innervated by multiple spiral ganglion neurons of type I (SGNI). SGNIs are morphologically and electro-physiologically diverse. Also, they differ in their susceptibility to noise insult. However, the molecular underpinnings of their identity and physiological differences remain poorly understood. In this study, we developed a novel triple transgenic mouse, which enabled the isolation of pure populations of SGNIs and the analysis of a 96-gene panel via single-cell qPCR. We found three distinct populations of Type I SGNs, which were marked by their exclusive expression of Lmx1a, Slc4a4, or Mfap4/Fzd2, respectively, at postnatal days P3, P8, and P12. Our data suggest that afferent SGN subtypes are established genetically before the onset of hearing and that the expression of key physiological markers, such as ion channels, is heterogeneous and may be underlying the heterogeneous firing proprieties of SGNIs.
Collapse
Affiliation(s)
| | - Lara De Tomasi
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Mirna Mustapha
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom.,Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
21
|
Bernard A, Chevrier S, Beltjens F, Dosset M, Viltard E, Lagrange A, Derangère V, Oudot A, Ghiringhelli F, Collin B, Apetoh L, Feron O, Chen S, Arnould L, Végran F, Boidot R. Cleaved Caspase-3 Transcriptionally Regulates Angiogenesis-Promoting Chemotherapy Resistance. Cancer Res 2019; 79:5958-5970. [PMID: 31611309 DOI: 10.1158/0008-5472.can-19-0840] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/17/2019] [Accepted: 10/08/2019] [Indexed: 11/16/2022]
Abstract
Caspases are well known for their role in apoptosis. Recently, nonapoptotic roles of caspases have been identified, however, these noncanonical roles are not well documented and the mechanisms involved are not fully understood. Here, we studied the role of cleaved caspase-3 using human- and mouse-proficient caspase-3 cancer cell lines and human-deficient caspase-3 cancer cells. Cleaved caspase-3 functioned as a transcription factor and directly bound to DNA. A DNA-binding domain was identified in the small subunit of caspase-3 and an active conformation was essential for caspase-3 transcriptional activity. Caspase-3 DNA binding enhanced angiogenesis by upregulating the expression of proangiogenic genes and by activating pathways that promoted endothelial cell activation. Some proapoptotic genes were downregulated in caspase-3-proficient cells. Inhibiting caspase-3 increased the efficacy of chemotherapy and decreased spontaneous tumor development. These data highlight a novel nonapoptotic role of caspase-3 and suggest that cleaved caspase-3 could be a new therapeutic target in cancer. SIGNIFICANCE: These findings report a noncanonical function of caspase-3 by demonstrating its ability to transcriptionally regulate the VEGFR pathway.
Collapse
Affiliation(s)
| | - Sandy Chevrier
- Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Françoise Beltjens
- Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | | | | | | | - Valentin Derangère
- Inserm U1231, Dijon, France.,Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Alexandra Oudot
- Preclinical Imaging Platform-Nuclear Medicine Department, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - François Ghiringhelli
- Inserm U1231, Dijon, France.,Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Bertrand Collin
- Preclinical Imaging Platform-Nuclear Medicine Department, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | | | - Olivier Feron
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Suzie Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey.,The Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Laurent Arnould
- Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Frédérique Végran
- Inserm U1231, Dijon, France.,Platform of Transfer in Cancer Biology, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Romain Boidot
- Inserm U1231, Dijon, France. .,Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| |
Collapse
|
22
|
Barber M, Andrews WD, Memi F, Gardener P, Ciantar D, Tata M, Ruhrberg C, Parnavelas JG. Vascular-Derived Vegfa Promotes Cortical Interneuron Migration and Proximity to the Vasculature in the Developing Forebrain. Cereb Cortex 2019; 28:2577-2593. [PMID: 29901792 PMCID: PMC5998991 DOI: 10.1093/cercor/bhy082] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Indexed: 12/29/2022] Open
Abstract
Vascular endothelial growth factor (Vegfa) is essential for promoting the vascularization of the embryonic murine forebrain. In addition, it directly influences neural development, although its role in the forming forebrain is less well elucidated. It was recently suggested that Vegfa may influence the development of GABAergic interneurons, inhibitory cells with crucial signaling roles in cortical neuronal circuits. However, the mechanism by which it affects interneuron development remains unknown. Here we investigated the developmental processes by which Vegfa may influence cortical interneuron development by analyzing transgenic mice that ubiquitously express the Vegfa120 isoform to perturb its signaling gradient. We found that interneurons reach the dorsal cortex at mid phases of corticogenesis despite an aberrant vascular network. Instead, endothelial ablation of Vegfa alters cortical interneuron numbers, their intracortical distribution and spatial proximity to blood vessels. We show for the first time that vascular-secreted guidance factors promote early-migrating interneurons in the intact forebrain in vivo and identify a novel role for vascular-Vegfa in this process.
Collapse
Affiliation(s)
- Melissa Barber
- Department of Cell and Developmental Biology, University College London, London, UK
| | - William D Andrews
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Fani Memi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Phillip Gardener
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Daniel Ciantar
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Mathew Tata
- Institute of Ophthalmology, University College London, London, UK
| | | | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
23
|
Taroc EZM, Lin JM, Tulloch AJ, Jaworski A, Forni PE. GnRH-1 Neural Migration From the Nose to the Brain Is Independent From Slit2, Robo3 and NELL2 Signaling. Front Cell Neurosci 2019; 13:70. [PMID: 30881290 PMCID: PMC6406018 DOI: 10.3389/fncel.2019.00070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/12/2019] [Indexed: 11/28/2022] Open
Abstract
Gonadotropin releasing hormone-1 (GnRH-1) neurons play a pivotal role in controlling pubertal onset and fertility once they reach their hypothalamic location. During embryonic development, GnRH-1 neurons migrate from the nasal area to the hypothalamus where they modulate gonadotropin release from the pituitary gland. Defective migration of the GnRH-1 neurons to the brain, lack of GnRH-1 secretion or signaling cause hypogonadotropic hypogonadism (HH), a pathology characterized by delayed or absence of puberty. Binding of the guidance cue Slit2 to the receptor roundabout 3 (Robo3) has been proposed to modulate GnRH-1 cell motility and basal forebrain (bFB) access during migration. However, evidence suggests that Neural EGFL Like 2 (NELL2), not Slit2, binds to Robo3. To resolve this discrepancy, we analyzed GnRH-1 neuronal migration in NELL2, Robo3, and Slit2 knock-out mouse lines. Our data do not confirm a negative effect for monogenic Robo3 and Slit2 mutations on GnRH-1 neuronal migration from the nasal area to the brain. Moreover, we found no changes in GnRH-1 neuronal migration in the brain after NELL2 loss-of-function. However, we found that Slit2 loss-of-function alters the patterning of GnRH-1 cells in the brain, suggesting that Slit2 loss-of-function affects GnRH-1 cell positioning in the brain in a Robo3 independent fashion. Our results challenge previous theories on GnRH-1 neuronal migration mechanisms and provide a new impetus to identify and understand the complex genetic mechanisms causing disorders like Kallmann syndrome (KS) and HH.
Collapse
Affiliation(s)
- Ed Zandro M Taroc
- Department of Biological Sciences, University at Albany, Albany, NY, United States
| | - Jennifer M Lin
- Department of Biological Sciences, University at Albany, Albany, NY, United States
| | - Alastair J Tulloch
- Department of Neuroscience, Brown University, Providence, RI, United States
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, RI, United States
| | - Paolo E Forni
- Department of Biological Sciences, University at Albany, Albany, NY, United States
| |
Collapse
|
24
|
Shadad O, Chaulagain R, Luukko K, Kettunen P. Establishment of tooth blood supply and innervation is developmentally regulated and takes place through differential patterning processes. J Anat 2019; 234:465-479. [PMID: 30793310 DOI: 10.1111/joa.12950] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2019] [Indexed: 01/08/2023] Open
Abstract
Teeth are richly supported by blood vessels and peripheral nerves. The aim of this study was to describe in detail the developmental time-course and localization of blood vessels during early tooth formation and to compare that to innervation, as well as to address the putative role of vascular endothelial growth factor (VEGF), which is an essential regulator of vasculature development, in this process. The localization of blood vessels and neurites was compared using double immunofluorescence staining on sections at consecutive stages of the embryonic (E) and postnatal (PN) mandibular first molar tooth germ (E11-PN7). Cellular mRNA expression domains of VEGF and its signaling receptor VEGFR2 were studied using sectional radioactive in situ hybridization. Expression of VEGF mRNA and the encoded protein were studied by RT-PCR and western blot analysis, respectively, in the cap and early bell stage tooth germs, respectively. VEGFR2 was immunolocalized on tooth tissue sections. Smooth muscle cells were investigated by anti-alpha smooth muscle actin (αSMA) antibodies. VEGF showed developmentally regulated epithelial and mesenchymal mRNA expression domains including the enamel knot signaling centers that correlated with the growth and navigation of the blood vessels expressing Vegfr2 and VEGFR2 to the dental papilla and enamel organ. Developing blood vessels were present in the jaw mesenchyme including the presumptive dental mesenchyme before the appearance of the epithelial dental placode and dental neurites. Similarly, formation of a blood vessel plexus around the bud stage tooth germ and ingrowth of vessels into dental papilla at E14 preceded ingrowth of neurites. Subsequently, pioneer blood vessels in the dental papilla started to receive smooth muscle coverage at the early embryonic bell stage. Establishment and patterning of the blood vessels and nerves during tooth formation are developmentally regulated, stepwise processes that likely involve differential patterning mechanisms. Development of tooth vascular supply is proposed to be regulated by local, tooth-specific regulation by epithelial-mesenchymal tissue interactions and involving tooth target expressed VEGF signaling. Further investigations on tooth vascular development by local VEGF signaling, as well as how tooth innervation and development of blood vessels are integrated with advancing tooth organ formation by local signaling mechanisms, are warranted.
Collapse
Affiliation(s)
- Omnia Shadad
- Department of Biomedicine, Craniofacial Developmental Biology Group, University of Bergen, Bergen, Norway.,Centre for International Health, University of Bergen, Bergen, Norway
| | - Rajib Chaulagain
- Department of Biomedicine, Craniofacial Developmental Biology Group, University of Bergen, Bergen, Norway.,Centre for International Health, University of Bergen, Bergen, Norway
| | - Keijo Luukko
- Department of Biomedicine, Craniofacial Developmental Biology Group, University of Bergen, Bergen, Norway.,Section of Orthodontics, Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Paivi Kettunen
- Department of Biomedicine, Craniofacial Developmental Biology Group, University of Bergen, Bergen, Norway
| |
Collapse
|
25
|
Memi F, Killen AC, Barber M, Parnavelas JG, Andrews WD. Cadherin 8 regulates proliferation of cortical interneuron progenitors. Brain Struct Funct 2018; 224:277-292. [PMID: 30315415 PMCID: PMC6373371 DOI: 10.1007/s00429-018-1772-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/05/2018] [Indexed: 01/18/2023]
Abstract
Cortical interneurons are born in the ventral forebrain and migrate tangentially in two streams at the levels of the intermediate zone (IZ) and the pre-plate/marginal zone to the developing cortex where they switch to radial migration before settling in their final positions in the cortical plate. In a previous attempt to identify the molecules that regulate stream specification, we performed transcriptomic analysis of GFP-labelled interneurons taken from the two migratory streams during corticogenesis. A number of cadherins were found to be expressed differentially, with Cadherin-8 (Cdh8) selectively present in the IZ stream. We verified this expression pattern at the mRNA and protein levels on tissue sections and found approximately half of the interneurons of the IZ expressed Cdh8. Furthermore, this cadherin was also detected in the germinal zones of the subpallium, suggesting that it might be involved not only in the migration of interneurons but also in their generation. Quantitative analysis of cortical interneurons in animals lacking the cadherin at E18.5 revealed a significant increase in their numbers. Subsequent functional in vitro experiments showed that blocking Cdh8 function led to increased cell proliferation, with the opposite results observed with over-expression, supporting its role in interneuron generation.
Collapse
Affiliation(s)
- Fani Memi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Abigail C Killen
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Melissa Barber
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - William D Andrews
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
27
|
Synaptic loss and firing alterations in Axotomized Motoneurons are restored by vascular endothelial growth factor (VEGF) and VEGF-B. Exp Neurol 2018. [PMID: 29522757 DOI: 10.1016/j.expneurol.2018.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vascular endothelial growth factor (VEGF), also known as VEGF-A, was discovered due to its vasculogenic and angiogenic activity, but a neuroprotective role for VEGF was later proven for lesions and disorders. In different models of motoneuronal degeneration, VEGF administration leads to a significant reduction of motoneuronal death. However, there is no information about the physiological state of spared motoneurons. We examined the trophic role of VEGF on axotomized motoneurons with recordings in alert animals using the oculomotor system as the experimental model, complemented with a synaptic study at the confocal microscopy level. Axotomy leads to drastic alterations in the discharge characteristics of abducens motoneurons, as well as to a substantial loss of their synaptic inputs. Retrograde delivery of VEGF completely restored the discharge activity and synaptically-driven signals in injured motoneurons, as demonstrated by correlating motoneuronal firing rate with motor performance. Moreover, VEGF-treated motoneurons recovered a normal density of synaptic boutons around motoneuronal somata and in the neuropil, in contrast to the low levels of synaptic terminals found after axotomy. VEGF also reduced the astrogliosis induced by axotomy in the abducens nucleus to control values. The administration of VEGF-B produced results similar to those of VEGF. This is the first work demonstrating that VEGF and VEGF-B restore the normal operating mode and synaptic inputs on injured motoneurons. Altogether these data indicate that these molecules are relevant synaptotrophic factors for motoneurons and support their clinical potential for the treatment of motoneuronal disorders.
Collapse
|
28
|
Characterization of neural stem cells modified with hypoxia/neuron-specific VEGF expression system for spinal cord injury. Gene Ther 2017; 25:27-38. [PMID: 29155421 DOI: 10.1038/gt.2017.92] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 09/30/2017] [Accepted: 10/04/2017] [Indexed: 02/01/2023]
Abstract
Spinal cord injury (SCI) is an incurable disease causing an ischemic environment and functional defect, thus a new therapeutic approach is needed for SCI treatment. Vascular endothelial growth factor (VEGF) is a potent therapeutic gene to treat SCI via angiogenesis and neuroprotection, and both tissue-specific gene expression and high gene delivery efficiency are important for successful gene therapy. Here we design the hypoxia/neuron dual-specific gene expression system (pEpo-NSE) and efficient gene delivery platform can be achieved by the combination ex vivo gene therapy with erythropoietin (Epo) enhancer, neuron-specific enolase (NSE) promoter and neural stem cells (NSCs). An in vitro model, NSCs transfected with pEpo-NSE were consistently and selectively overexpressing therapeutic genes in response to neural differentiation and hypoxic conditions. Also, in SCI model, ex vivo gene therapy using pEpo-NSE system with NSCs significantly enhanced gene delivery efficiency compared with pEpo-NSE system gene therapy alone. However, microarray analysis reveals that introducing exogenous pEpo-NSE and VEGF triggers biological pathways in NSCs such as glycolysis and signaling pathways such as Ras and mitogen-activated protein kinase, leading to cell proliferation, differentiation and apoptosis. Collectively, it indicates that the pEpo-NSE gene expression system works stably in NSCs and ex vivo gene therapy using pEpo-NSE system with NSCs improves gene expression efficiency. However, exogenously introduced pEpo-NSE system has an influence on gene expression profiles in NSCs. Therefore, when we consider ex vivo gene therapy for SCI, the effects of changes in gene expression profiles in NSCs on safety should be investigated.
Collapse
|
29
|
Salehi P, Ge MX, Gundimeda U, Michelle Baum L, Lael Cantu H, Lavinsky J, Tao L, Myint A, Cruz C, Wang J, Nikolakopoulou AM, Abdala C, Kelley MW, Ohyama T, Coate TM, Friedman RA. Role of Neuropilin-1/Semaphorin-3A signaling in the functional and morphological integrity of the cochlea. PLoS Genet 2017; 13:e1007048. [PMID: 29059194 PMCID: PMC5695633 DOI: 10.1371/journal.pgen.1007048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 11/02/2017] [Accepted: 09/28/2017] [Indexed: 11/18/2022] Open
Abstract
Neuropilin-1 (Nrp1) encodes the transmembrane cellular receptor neuropilin-1, which is associated with cardiovascular and neuronal development and was within the peak SNP interval on chromosome 8 in our prior GWAS study on age-related hearing loss (ARHL) in mice. In this study, we generated and characterized an inner ear-specific Nrp1 conditional knockout (CKO) mouse line because Nrp1 constitutive knockouts are embryonic lethal. In situ hybridization demonstrated weak Nrp1 mRNA expression late in embryonic cochlear development, but increased expression in early postnatal stages when cochlear hair cell innervation patterns have been shown to mature. At postnatal day 5, Nrp1 CKO mice showed disorganized outer spiral bundles and enlarged microvessels of the stria vascularis (SV) but normal spiral ganglion cell (SGN) density and presynaptic ribbon body counts; however, we observed enlarged SV microvessels, reduced SGN density, and a reduction of presynaptic ribbons in the outer hair cell region of 4-month-old Nrp1 CKO mice. In addition, we demonstrated elevated hearing thresholds of the 2-month-old and 4-month-old Nrp1 CKO mice at frequencies ranging from 4 to 32kHz when compared to 2-month-old mice. These data suggest that conditional loss of Nrp1 in the inner ear leads to progressive hearing loss in mice. We also demonstrated that mice with a truncated variant of Nrp1 show cochlear axon guidance defects and that exogenous semaphorin-3A, a known neuropilin-1 receptor agonist, repels SGN axons in vitro. These data suggest that Neuropilin-1/Semaphorin-3A signaling may also serve a role in neuronal pathfinding in the developing cochlea. In summary, our results here support a model whereby Neuropilin-1/Semaphorin-3A signaling is critical for the functional and morphological integrity of the cochlea and that Nrp1 may play a role in ARHL. Neuropilin-1 is a member of the neuropilin family acting as an essential cell surface receptor involved in semaphorin-dependent axon guidance and VEGF-dependent angiogenesis and lies within our previously identified ARHL GWAS interval. In this study, we investigated the role of Neuropilin-1/Semaphorin-3A signaling in the functional and morphological integrity of the cochlea, specifically the innervation and vascularization patterns. Detailed analyses of the cochleae of 4-month-old Nrp1 CKO mice showed abnormalities in ribbon synapses, innervation of the hair cells, and microvessels of the stria vascularis. We show also that Neuropilin-1/Semaphorin-3A signaling plays an important role in cochlear innervation.
Collapse
Affiliation(s)
- Pezhman Salehi
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, United States of America
| | - Marshall X. Ge
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Usha Gundimeda
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Leah Michelle Baum
- Department of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Homero Lael Cantu
- Department of Biology, Georgetown University, Washington, D.C., United States of America
| | - Joel Lavinsky
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Graduate Program in Surgical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Litao Tao
- Stem Cell Biology & Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Anthony Myint
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Charlene Cruz
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Juemei Wang
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Carolina Abdala
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Matthew William Kelley
- National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland, United States of America
| | - Takahiro Ohyama
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Thomas Matthew Coate
- Department of Biology, Georgetown University, Washington, D.C., United States of America
- * E-mail: (TMC); (RAF)
| | - Rick A. Friedman
- USC-Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Zilkha Neurogenetic Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail: (TMC); (RAF)
| |
Collapse
|
30
|
Erskine L, François U, Denti L, Joyce A, Tillo M, Bruce F, Vargesson N, Ruhrberg C. VEGF-A and neuropilin 1 (NRP1) shape axon projections in the developing CNS via dual roles in neurons and blood vessels. Development 2017; 144:2504-2516. [PMID: 28676569 PMCID: PMC5536872 DOI: 10.1242/dev.151621] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
Abstract
Visual information is relayed from the eye to the brain via retinal ganglion cell (RGC) axons. Mice lacking NRP1 or NRP1-binding VEGF-A isoforms have defective RGC axon organisation alongside brain vascular defects. It is not known whether axonal defects are caused exclusively by defective VEGF-A signalling in RGCs or are exacerbated by abnormal vascular morphology. Targeted NRP1 ablation in RGCs with a Brn3bCre knock-in allele reduced axonal midline crossing at the optic chiasm and optic tract fasciculation. In contrast, Tie2-Cre-mediated endothelial NRP1 ablation induced axon exclusion zones in the optic tracts without impairing axon crossing. Similar defects were observed in Vegfa120/120 and Vegfa188/188 mice, which have vascular defects as a result of their expression of single VEGF-A isoforms. Ectopic midline vascularisation in endothelial Nrp1 and Vegfa188/188 mutants caused additional axonal exclusion zones within the chiasm. As in vitro and in vivo assays demonstrated that vessels do not repel axons, abnormally large or ectopically positioned vessels are likely to present physical obstacles to axon growth. We conclude that proper axonal wiring during brain development depends on the precise molecular control of neurovascular co-patterning. Summary: NRP1 plays a dual role in retinal ganglion cells and in vascular endothelial cells to organise axons along the optic pathway between the mouse retina and diencephalon.
Collapse
Affiliation(s)
- Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Urielle François
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Andy Joyce
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Miguel Tillo
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Freyja Bruce
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
31
|
Liu Y, Yan Y, Inagaki Y, Logan S, Bosnjak ZJ, Bai X. Insufficient Astrocyte-Derived Brain-Derived Neurotrophic Factor Contributes to Propofol-Induced Neuron Death Through Akt/Glycogen Synthase Kinase 3β/Mitochondrial Fission Pathway. Anesth Analg 2017. [PMID: 28622174 DOI: 10.1213/ane.0000000000002137] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Growing animal evidence demonstrates that prolonged exposure to propofol during brain development induces widespread neuronal cell death, but there is little information on the role of astrocytes. Astrocytes can release neurotrophic growth factors such as brain-derived neurotrophic factor (BDNF), which can exert the protective effect on neurons in paracrine fashion. We hypothesize that during propofol anesthesia, BDNF released from developing astrocytes may not be sufficient to prevent propofol-induced neurotoxicity. METHODS Hippocampal astrocytes and neurons isolated from neonatal Sprague Dawley rats were exposed to propofol at a clinically relevant dose of 30 μM or dimethyl sulfoxide as control for 6 hours. Propofol-induced cell death was determined by propidium iodide (PI) staining in astrocyte-alone cultures, neuron-alone cultures, or cocultures containing either low or high density of astrocytes (1:9 or 1:1 ratio of astrocytes to neurons ratio [ANR], respectively). The astrocyte-conditioned medium was collected 12 hours after propofol exposure and measured by protein array assay. BDNF concentration in astrocyte-conditioned medium was quantified using enzyme-linked immunosorbent assay. Neuron-alone cultures were treated with BDNF, tyrosine receptor kinase B inhibitor cyclotraxin-B, glycogen synthase kinase 3β (GSK3β) inhibitor CHIR99021, or mitochondrial fission inhibitor Mdivi-1 before propofol exposure. Western blot was performed for quantification of the level of protein kinase B and GSK3β. Mitochondrial shape was visualized through translocase of the outer membrane 20 staining. RESULTS Propofol increased cell death in neurons by 1.8-fold (% of PI-positive cells [PI%] = 18.6; 95% confidence interval [CI], 15.2-21.9, P < .05) but did not influence astrocyte viability. The neuronal death was attenuated by a high ANR (1:1 cocultures; fold change [FC] = 1.17, 95% CI, 0.96-1.38, P < .05), but not with a low ANR [1:9 cocultures; FC = 1.87, 95% CI, 1.48-2.26, P > .05]). Astrocytes secreted BDNF in a cell density-dependent way and propofol decreased BDNF secretion from astrocytes. Administration of BDNF, CHIR99021, or Mdivi-1 significantly attenuated the propofol-induced neuronal death and aberrant mitochondria in neuron-alone cultures (FC = 0.8, 95% CI, 0.62-0.98; FC = 1.22, 95% CI, 1.11-1.32; FC = 1.35, 95% CI, 1.16-1.54, respectively, P < .05) and the cocultures with a low ANR (1:9; FC = 0.85, 95% CI, 0.74-0.97; FC = 1.08, 95% CI, 0.84-1.32; FC = 1.25, 95% CI, 1.1-1.39, respectively, P < .05). Blocking BDNF receptor or protein kinase B activity abolished astrocyte-induced neuroprotection in the cocultures with a high ANR (1:1). CONCLUSIONS Astrocytes attenuate propofol-induced neurotoxicity through BDNF-mediated cell survival pathway suggesting multiple neuroprotective strategies such as administration of BDNF, astrocyte-conditioned medium, decreasing mitochondrial fission, or inhibition of GSK3β.
Collapse
Affiliation(s)
- Yanan Liu
- From the Departments of *Anesthesiology and †Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | | | | | | | | |
Collapse
|
32
|
Killen AC, Barber M, Paulin JJW, Ranscht B, Parnavelas JG, Andrews WD. Protective role of Cadherin 13 in interneuron development. Brain Struct Funct 2017; 222:3567-3585. [PMID: 28386779 PMCID: PMC5676827 DOI: 10.1007/s00429-017-1418-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/30/2017] [Indexed: 12/21/2022]
Abstract
Cortical interneurons are generated in the ganglionic eminences and migrate through the ventral and dorsal telencephalon before finding their final positions within the cortical plate. During early stages of migration, these cells are present in two well-defined streams within the developing cortex. In an attempt to identify candidate genes which may play a role in interneuron stream specification, we previously carried out a microarray analysis which identified a number of cadherin receptors that were differentially expressed in these streams, including Cadherin-13 (Cdh13). Expression analysis confirmed Cdh13 to be present in the preplate layer at E13.5 and, later in development, in some cortical interneurons and pyramidal cells. Analysis of Cdh13 knockout mice at E18.5, but not at E15.5, showed a reduction in the number of interneurons and late born pyramidal neurons and a concomitant increase in apoptotic cells in the cortex. These observations were confirmed in dissociated cell cultures using overexpression and short interfering RNAs (siRNAs) constructs and dominant negative inhibitory proteins. Our findings identified a novel protective role for Cdh13 in cortical neuron development.
Collapse
Affiliation(s)
- Abigail C Killen
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Melissa Barber
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Joshua J W Paulin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Barbara Ranscht
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - John G Parnavelas
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - William D Andrews
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
33
|
Fantin A, Lampropoulou A, Senatore V, Brash JT, Prahst C, Lange CA, Liyanage SE, Raimondi C, Bainbridge JW, Augustin HG, Ruhrberg C. VEGF165-induced vascular permeability requires NRP1 for ABL-mediated SRC family kinase activation. J Exp Med 2017; 214:1049-1064. [PMID: 28289053 PMCID: PMC5379968 DOI: 10.1084/jem.20160311] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 12/07/2016] [Accepted: 01/20/2017] [Indexed: 01/13/2023] Open
Abstract
Fantin et al. show that the VEGF isoform VEGF165 signals through a complex of VEGFR2 and NRP1, in which the NRP1 cytoplasmic domain promotes the ABL-mediated activation of SRC family kinases to evoke a hyperpermeability response, a known cause of pathological edema. The vascular endothelial growth factor (VEGF) isoform VEGF165 stimulates vascular growth and hyperpermeability. Whereas blood vessel growth is essential to sustain organ health, chronic hyperpermeability causes damaging tissue edema. By combining in vivo and tissue culture models, we show here that VEGF165-induced vascular leakage requires both VEGFR2 and NRP1, including the VEGF164-binding site of NRP1 and the NRP1 cytoplasmic domain (NCD), but not the known NCD interactor GIPC1. In the VEGF165-bound receptor complex, the NCD promotes ABL kinase activation, which in turn is required to activate VEGFR2-recruited SRC family kinases (SFKs). These results elucidate the receptor complex and signaling hierarchy of downstream kinases that transduce the permeability response to VEGF165. In a mouse model with choroidal neovascularisation akin to age-related macular degeneration, NCD loss attenuated vessel leakage without affecting neovascularisation. These findings raise the possibility that targeting NRP1 or its NCD interactors may be a useful therapeutic strategy in neovascular disease to reduce VEGF165-induced edema without compromising vessel growth.
Collapse
Affiliation(s)
- Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | | | - Valentina Senatore
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Claudia Prahst
- Yale Cardiovascular Research Center, New Haven, CT 06511
| | - Clemens A Lange
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Sidath E Liyanage
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - James W Bainbridge
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| |
Collapse
|
34
|
Liu F, Jiao Y, Jiao Y, Garcia-Godoy F, Gu W, Liu Q. Sex difference in EGFR pathways in mouse kidney-potential impact on the immune system. BMC Genet 2016; 17:146. [PMID: 27881077 PMCID: PMC5122204 DOI: 10.1186/s12863-016-0449-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/15/2016] [Indexed: 12/23/2022] Open
Abstract
Background Epidermal growth factor receptor (Egfr) has been the target of several drugs for cancers. The potential gender differences in genes in the Egfr axis have been suggested in humans and in animal models. Female and male mice from the same recombinant inbred (RI) strain have the same genomic components except the sex difference. A population of different RI mouse strains allows to conduct precise analysis of molecular pathways and regulation of Egfr between female and male mice. Methods The whole genome expression profiles of 70 genetically diverse RI strains of mice were used to compare three major molecular aspects of Egfr gene: the relative expression levels, gene network and expression quantitative trait loci (eQTL) that regulate the expression of Egfr between female and male mice. Results Our data showed that there is a significant sex difference in the expression levels in kidney. A considerable number of genes in the gene network of Egfr are sex differentially expressed. The expression levels of Egfr in mice are statistical significant different between C57BL/6 J (B6) and DBA/2 J (D2) genotypes in male while no difference in female mice. The eQTLs that regulate the expression levels of Egfr between female and male mice are also different. Furthermore, the differential expression levels of Egfr showed significantly different correlations with two known biological traits between male and female mice. Conclusion Overall there is a substantial sex difference in the Egfr pathways in mice. These data may have significant impact on drug target design, development, formulation, and dosage determinant for women and men in clinical trials. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0449-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fengxia Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.,Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yan Jiao
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yun Jiao
- Department of Neuroscience, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Franklin Garcia-Godoy
- Bioscience Research Center, College of Dentistry, University of Tennessee Health Science Center, 875 Union Avenue, Memphis, TN, USA
| | - Weikuan Gu
- Department of Orthopaedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN, USA. .,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, USA. .,, 956 Court Ave, Memphis, TN, 38163, USA.
| | - Qingyi Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei, 050011, China.
| |
Collapse
|
35
|
Xu D, Chen X, He Q, Luo C. MicroRNA-9 suppresses the growth, migration, and invasion of malignant melanoma cells via targeting NRP1. Onco Targets Ther 2016; 9:7047-7057. [PMID: 27895497 PMCID: PMC5117879 DOI: 10.2147/ott.s107235] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRs) are a class of small noncoding RNAs that negatively regulate the gene expression by directly binding to the 3' untranslated region of their target mRNA, thus resulting in mRNA degradation or translational repression. miR-9 has recently been demonstrated to play a role in the development and progression of malignant melanoma (MM), but the regulatory mechanism of miR-9 in the malignant phenotypes of MM still remains largely unknown. In this study, a total of 73 pairs of MM tissues and adjacent normal tissues were collected. Real-time reverse transcription polymerase chain reaction and Western blot were used to detect the mRNA and protein expression of miR-9. MTT assay, wound healing assay, and transwell assay were conducted to determine the cell proliferation, migration, and invasion. Luciferase reporter assay was used to determine the targeting relationship between miR-9 and NRP1. Our data demonstrated that miR-9 expression was significantly downregulated in MM tissues compared with that in adjacent normal tissues. The decreased miR-9 level was significantly associated with the tumor stage and metastasis of MM. We also found that the expression level of miR-9 was decreased in MM cell lines (G361, B16, A375, and HME1) compared with normal skin HACAT cells. Ectopic expression of miR-9 led to a significant decrease in the ability of proliferation, migration, and invasion in A375 cells. NRP1 was further identified as a direct target gene of miR-9, and the protein expression of NRP1 was negatively regulated by miR-9 in A375 cells. Furthermore, overexpression of NRP1 reversed the suppressive effects of miR-9 on the malignant phenotypes of A375 cells. In vivo study revealed that miR-9 overexpression decreased the tumor growth, while overexpression of NRP1 increased MM growth. In summary, our findings suggest that the miR-9/NRP1 axis may serve as a potential target for the treatment of MM.
Collapse
Affiliation(s)
- Dan Xu
- Department of Plastic Surgery, Third Xiangya Hospital of Central South University
| | - Xiaofeng Chen
- Department of Neurosurgery, Brain Hospital of Hunan Province, Changsha, Hunan, People's Republic of China
| | - Quanyong He
- Department of Plastic Surgery, Third Xiangya Hospital of Central South University
| | - Chengqun Luo
- Department of Plastic Surgery, Third Xiangya Hospital of Central South University
| |
Collapse
|
36
|
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are uniquely required to balance the formation of new blood vessels with the maintenance and remodelling of existing ones, during development and in adult tissues. Recent advances have greatly expanded our understanding of the tight and multi-level regulation of VEGFR2 signalling, which is the primary focus of this Review. Important insights have been gained into the regulatory roles of VEGFR-interacting proteins (such as neuropilins, proteoglycans, integrins and protein tyrosine phosphatases); the dynamics of VEGFR2 endocytosis, trafficking and signalling; and the crosstalk between VEGF-induced signalling and other endothelial signalling cascades. A clear understanding of this multifaceted signalling web is key to successful therapeutic suppression or stimulation of vascular growth.
Collapse
|
37
|
Howard SR, Guasti L, Ruiz-Babot G, Mancini A, David A, Storr HL, Metherell LA, Sternberg MJ, Cabrera CP, Warren HR, Barnes MR, Quinton R, de Roux N, Young J, Guiochon-Mantel A, Wehkalampi K, André V, Gothilf Y, Cariboni A, Dunkel L. IGSF10 mutations dysregulate gonadotropin-releasing hormone neuronal migration resulting in delayed puberty. EMBO Mol Med 2016; 8:626-42. [PMID: 27137492 PMCID: PMC4888853 DOI: 10.15252/emmm.201606250] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Early or late pubertal onset affects up to 5% of adolescents and is associated with adverse health and psychosocial outcomes. Self‐limited delayed puberty (DP) segregates predominantly in an autosomal dominant pattern, but the underlying genetic background is unknown. Using exome and candidate gene sequencing, we have identified rare mutations in IGSF10 in 6 unrelated families, which resulted in intracellular retention with failure in the secretion of mutant proteins. IGSF10 mRNA was strongly expressed in embryonic nasal mesenchyme, during gonadotropin‐releasing hormone (GnRH) neuronal migration to the hypothalamus. IGSF10 knockdown caused a reduced migration of immature GnRH neurons in vitro, and perturbed migration and extension of GnRH neurons in a gnrh3:EGFP zebrafish model. Additionally, loss‐of‐function mutations in IGSF10 were identified in hypothalamic amenorrhea patients. Our evidence strongly suggests that mutations in IGSF10 cause DP in humans, and points to a common genetic basis for conditions of functional hypogonadotropic hypogonadism (HH). While dysregulation of GnRH neuronal migration is known to cause permanent HH, this is the first time that this has been demonstrated as a causal mechanism in DP.‡
Collapse
Affiliation(s)
- Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gerard Ruiz-Babot
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alessandra Mancini
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alessia David
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, UK
| | - Helen L Storr
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Lousie A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Je Sternberg
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, UK
| | - Claudia P Cabrera
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Helen R Warren
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Michael R Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Richard Quinton
- Institute of Genetic Medicine University of Newcastle-upon-Tyne, Newcastle-upon-Tyne, UK
| | - Nicolas de Roux
- Unité Mixte de Recherche 1141, Institut National de la Santé et de la Recherche Médicale, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France Laboratoire de Biochimie, Assistance Publique-Hôpitaux de Paris, Hôpital Robert Debré, Paris, France
| | - Jacques Young
- Univ Paris-Sud, Le Kremlin Bicêtre, France INSERM UMR-1185, Le Kremlin Bicêtre, France Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France Department of Reproductive Endocrinology, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Anne Guiochon-Mantel
- Univ Paris-Sud, Le Kremlin Bicêtre, France INSERM UMR-1185, Le Kremlin Bicêtre, France Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Karoliina Wehkalampi
- Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Valentina André
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Yoav Gothilf
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences and Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy Institute of Ophthalmology, University College London (UCL), London, UK
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
38
|
Foxton R, Osborne A, Martin KR, Ng YS, Shima DT. Distal retinal ganglion cell axon transport loss and activation of p38 MAPK stress pathway following VEGF-A antagonism. Cell Death Dis 2016; 7:e2212. [PMID: 27148685 PMCID: PMC4917649 DOI: 10.1038/cddis.2016.110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/04/2016] [Accepted: 03/16/2016] [Indexed: 01/10/2023]
Abstract
There is increasing evidence that VEGF-A antagonists may be detrimental to neuronal health following ocular administration. Here we investigated firstly the effects of VEGF-A neutralization on retinal neuronal survival in the Ins2Akita diabetic and JR5558 spontaneous choroidal neovascularization (CNV) mice, and then looked at potential mechanisms contributing to cell death. We detected elevated apoptosis in the ganglion cell layer in both these models following VEGF-A antagonism, indicating that even when vascular pathologies respond to treatment, neurons are still vulnerable to reduced VEGF-A levels. We observed that retinal ganglion cells (RGCs) seemed to be the cells most susceptible to VEGF-A antagonism, so we looked at anterograde transport in these cells, due to their long axons requiring optimal protein and organelle trafficking. Using cholera toxin B-subunit tracer studies, we found a distal reduction in transport in the superior colliculus following VEGF-A neutralization, which occurred prior to net RGC loss. This phenomenon of distal transport loss has been described as a feature of early pathological changes in glaucoma, Alzheimer's and Parkinson's disease models. Furthermore, we observed increased phosphorylation of p38 MAPK and downstream Hsp27 stress pathway signaling in the retinas from these experiments, potentially providing a mechanistic explanation for our findings. These experiments further highlight the possible risks of using VEGF-A antagonists to treat ocular neovascular disease, and suggest that VEGF-A may contribute to the maintenance and function of axonal transport in neurons of the retina.
Collapse
Affiliation(s)
- R Foxton
- Department of Ocular Biology and Therapeutics, University College London, Institute of Ophthalmology, London, UK
| | - A Osborne
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - K R Martin
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK.,Wellcome Trust Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK.,Cambridge NIHR Biomedical Research Centre, Cambridge, UK.,Eye Department, Addenbrooke's Hospital, Cambridge, UK
| | - Y-S Ng
- Department of Ocular Biology and Therapeutics, University College London, Institute of Ophthalmology, London, UK.,The Schepens Eye Research Institute and Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - D T Shima
- Department of Ocular Biology and Therapeutics, University College London, Institute of Ophthalmology, London, UK
| |
Collapse
|
39
|
Latzer P, Schlegel U, Theiss C. Morphological Changes of Cortical and Hippocampal Neurons after Treatment with VEGF and Bevacizumab. CNS Neurosci Ther 2016; 22:440-50. [PMID: 26861512 PMCID: PMC5067574 DOI: 10.1111/cns.12516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/21/2015] [Accepted: 01/02/2016] [Indexed: 01/13/2023] Open
Abstract
Aims Vascular endothelial growth factor (VEGF) is a hallmark of glioblastoma multiforme (GBM) and plays an important role in brain development and function. Recently, it has been reported that treatment of GBM patients with bevacizumab, an anti‐VEGF antibody, may cause a decline in neurocognitive function and compromise quality of life. Therefore, we investigated the effects of VEGF and bevacizumab on the morphology and on survival of neurons and glial cells. Methods Dissociated cortical and hippocampal cell cultures of juvenile rats were treated with VEGF, bevacizumab, and VEGF + bevacizumab. Neuronal and glial cell viability was analyzed, and the morphology of neurons was objectified by morphometric analysis. Results In cortical cultures, bevacizumab significantly decreased the number of neurons after 20 days and the number of glial cells subsequent 30 days. Additionally, an increase in the dendritic length of cortical neurons was obvious after 10 days of incubation with bevacizumab, but returned to control level after 30 days. In hippocampal cultures, cell viability was not affected by bevacizumab; however, dendritic length increased at day 10, but decreased after long‐term treatment. Conclusion Therefore, bevacizumab obviously has a cytotoxic effect in cortical cultures and decreases the dendritic length in hippocampal neurons after long‐term treatment.
Collapse
Affiliation(s)
- Pauline Latzer
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Uwe Schlegel
- Department of Neurology, Knappschaftskrankenhaus, Ruhr University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
40
|
Abstract
The developing central nervous system (CNS) is vascularised through the angiogenic invasion of blood vessels from a perineural vascular plexus, followed by continued sprouting and remodelling until a hierarchical vascular network is formed. Remarkably, vascularisation occurs without perturbing the intricate architecture of the neurogenic niches or the emerging neural networks. We discuss the mouse hindbrain, forebrain and retina as widely used models to study developmental angiogenesis in the mammalian CNS and provide an overview of key cellular and molecular mechanisms regulating the vascularisation of these organs. CNS vascularisation is initiated during embryonic development. CNS vascularisation is studied in the mouse forebrain, hindbrain and retina models. Neuroglial cells interact with endothelial cells to promote angiogenesis. Neuroglial cells produce growth factors and matrix cues to pattern vessels.
Collapse
|
41
|
Cariboni A, André V, Chauvet S, Cassatella D, Davidson K, Caramello A, Fantin A, Bouloux P, Mann F, Ruhrberg C. Dysfunctional SEMA3E signaling underlies gonadotropin-releasing hormone neuron deficiency in Kallmann syndrome. J Clin Invest 2015; 125:2413-28. [PMID: 25985275 DOI: 10.1172/jci78448] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 04/02/2015] [Indexed: 01/09/2023] Open
Abstract
Individuals with an inherited deficiency in gonadotropin-releasing hormone (GnRH) have impaired sexual reproduction. Previous genetic linkage studies and sequencing of plausible gene candidates have identified mutations associated with inherited GnRH deficiency, but the small number of affected families and limited success in validating candidates have impeded genetic diagnoses for most patients. Using a combination of exome sequencing and computational modeling, we have identified a shared point mutation in semaphorin 3E (SEMA3E) in 2 brothers with Kallmann syndrome (KS), which causes inherited GnRH deficiency. Recombinant wild-type SEMA3E protected maturing GnRH neurons from cell death by triggering a plexin D1-dependent (PLXND1-dependent) activation of PI3K-mediated survival signaling. In contrast, recombinant SEMA3E carrying the KS-associated mutation did not protect GnRH neurons from death. In murine models, lack of either SEMA3E or PLXND1 increased apoptosis of GnRH neurons in the developing brain, reducing innervation of the adult median eminence by GnRH-positive neurites. GnRH neuron deficiency in male mice was accompanied by impaired testes growth, a characteristic feature of KS. Together, these results identify SEMA3E as an essential gene for GnRH neuron development, uncover a neurotrophic function for SEMA3E in the developing brain, and elucidate SEMA3E/PLXND1/PI3K signaling as a mechanism that prevents GnRH neuron deficiency.
Collapse
|
42
|
Plein A, Fantin A, Ruhrberg C. Neuropilin regulation of angiogenesis, arteriogenesis, and vascular permeability. Microcirculation 2015; 21:315-23. [PMID: 24521511 PMCID: PMC4230468 DOI: 10.1111/micc.12124] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
Abstract
The formation of the cardiovasculature, consisting of both the heart and blood vessels, is a critical step in embryonic development and relies on three processes termed vasculogenesis, angiogenesis, and vascular remodeling. The transmembrane protein NRP1 is an essential modulator of embryonic angiogenesis with additional roles in vessel remodeling and arteriogenesis. NRP1 also enhances arteriogenesis in adults to alleviate pathological tissue ischemia. However, in certain circumstances, vascular NRP1 signaling can be detrimental, as it may promote cancer by enhancing tumor angiogenesis or contribute to tissue edema by increasing vascular permeability. Understanding the mechanisms of NRP1 signaling is, therefore, of profound importance for the design of therapies aiming to control vascular functions. Previous work has shown that vascular NRP1 can variably serve as a receptor for two secreted glycoproteins, the VEGF-A and SEMA3A, but it also has a poorly understood role as an adhesion receptor. Here, we review current knowledge of NRP1 function during blood vessel growth and homeostasis, with special emphasis on the vascular roles of its multiple ligands and signaling partners.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
43
|
Giacobini P. Shaping the Reproductive System: Role of Semaphorins in Gonadotropin-Releasing Hormone Development and Function. Neuroendocrinology 2015; 102:200-15. [PMID: 25967979 DOI: 10.1159/000431021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/28/2015] [Indexed: 11/19/2022]
Abstract
The semaphorin proteins, which contribute to the morphogenesis and homeostasis of a wide range of systems, are among the best-studied families of guidance cues. Much recent research has focused on the role of semaphorins in the development and adult activity of hormone systems and, reciprocally, how circulating reproductive hormones regulate their expression and function. Specifically, several reports have focused on the molecular mechanisms underlying the effects of semaphorins on the migration, survival and structural and functional plasticity of neurons that secrete gonadotropin-releasing hormone (GnRH), essential for the acquisition and maintenance of reproductive competence in mammals. Alterations in the development of this neuroendocrine system lead to anomalous or absent GnRH secretion, resulting in heterogeneous reproductive disorders such as congenital hypogonadotropic hypogonadism (CHH) or other conditions characterized by infertility or subfertility. This review summarizes current knowledge of the role of semaphorins and their receptors on the development, differentiation and plasticity of the GnRH system. In addition, the involvement of genetic deficits in semaphorin signaling in some forms of CHH in humans is discussed.
Collapse
Affiliation(s)
- Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, School of Medicine, University of Lille, and Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| |
Collapse
|
44
|
Tillo M, Erskine L, Cariboni A, Fantin A, Joyce A, Denti L, Ruhrberg C. VEGF189 binds NRP1 and is sufficient for VEGF/NRP1-dependent neuronal patterning in the developing brain. Development 2014; 142:314-9. [PMID: 25519242 PMCID: PMC4302834 DOI: 10.1242/dev.115998] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The vascular endothelial growth factor (VEGFA, VEGF) regulates neurovascular patterning. Alternative splicing of the Vegfa gene gives rise to three major isoforms termed VEGF121, VEGF165 and VEGF189. VEGF165 binds the transmembrane protein neuropilin 1 (NRP1) and promotes the migration, survival and axon guidance of subsets of neurons, whereas VEGF121 cannot activate NRP1-dependent neuronal responses. By contrast, the role of VEGF189 in NRP1-mediated signalling pathways has not yet been examined. Here, we have combined expression studies and in situ ligand-binding assays with the analysis of genetically altered mice and in vitro models to demonstrate that VEGF189 can bind NRP1 and promote NRP1-dependent neuronal responses. Summary: Although VEGF165 was thought to be the sole VEGF isoform acting through neuropilin 1 (NRP1), VEGF189 also binds to and signals through NRP1 in several types of developing mouse neurons.
Collapse
Affiliation(s)
- Miguel Tillo
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Lynda Erskine
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Anna Cariboni
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK University of Milan, Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, Milan 20133, Italy
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Andy Joyce
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
45
|
Giacobini P, Parkash J, Campagne C, Messina A, Casoni F, Vanacker C, Langlet F, Hobo B, Cagnoni G, Gallet S, Hanchate NK, Mazur D, Taniguchi M, Mazzone M, Verhaagen J, Ciofi P, Bouret SG, Tamagnone L, Prevot V. Brain endothelial cells control fertility through ovarian-steroid-dependent release of semaphorin 3A. PLoS Biol 2014; 12:e1001808. [PMID: 24618750 PMCID: PMC3949669 DOI: 10.1371/journal.pbio.1001808] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/30/2014] [Indexed: 11/25/2022] Open
Abstract
Endothelial-cell–derived Sema3A promotes axonal outgrowth and plasticity and thereby regulates neurohormone release in the adult rodent brain in response to the ovarian cycle. Neuropilin-1 (Nrp1) guides the development of the nervous and vascular systems, but its role in the mature brain remains to be explored. Here we report that the expression of the 65 kDa isoform of Sema3A, the ligand of Nrp1, by adult vascular endothelial cells, is regulated during the ovarian cycle and promotes axonal sprouting in hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH), the neuropeptide controlling reproduction. Both the inhibition of Sema3A/Nrp1 signaling and the conditional deletion of Nrp1 in GnRH neurons counteract Sema3A-induced axonal sprouting. Furthermore, the localized intracerebral infusion of Nrp1- or Sema3A-neutralizing antibodies in vivo disrupts the ovarian cycle. Finally, the selective neutralization of endothelial-cell Sema3A signaling in adult Sema3aloxP/loxP mice by the intravenous injection of the recombinant TAT-Cre protein alters the amplitude of the preovulatory luteinizing hormone surge, likely by perturbing GnRH release into the hypothalamo-hypophyseal portal system. Our results identify a previously unknown function for 65 kDa Sema3A-Nrp1 signaling in the induction of axonal growth, and raise the possibility that endothelial cells actively participate in synaptic plasticity in specific functional domains of the adult central nervous system, thus controlling key physiological functions such as reproduction. In the developing embryo, endothelial cells release chemotropic signals such as Semaphorin 3A (Sema3A) that, upon activation of its receptor Neuropilin-1 (Nrp1), regulate neuronal migration and axon guidance. However, whether endothelial cells in the adult brain retain the ability to secrete molecules that influence neuronal function is unknown. Here we show in the adult brain of rodents that vascular endothelial cells release Sema3A and that the amount released is regulated by the ovulatory cycle. Sema3A, in turn, promotes the outgrowth of axons of hypothalamic neurons that express Neuropilin-1 towards the endothelial wall of portal blood vessels. These neurons release there the neuropeptide that controls reproduction: gonadotropin-releasing hormone (GnRH). Notably, this endothelial-cell-mediated sprouting of GnRH axons regulates neuropeptide release at a key stage of the estrous cycle, the proestrus, when the surge of GnRH triggers ovulation. Thus, by promoting GnRH axonal growth in the adult brain, Sema3A/Neuropilin-1 plays a pivotal role in orchestrating the central control of reproduction. Our results suggest a model in which vascular endothelial cells are dynamic signaling components that relay peripheral information to the brain to control key physiological functions, including species survival.
Collapse
Affiliation(s)
- Paolo Giacobini
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Jyoti Parkash
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Céline Campagne
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Andrea Messina
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Filippo Casoni
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Charlotte Vanacker
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Fanny Langlet
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Barbara Hobo
- Netherlands institute for Neuroscience, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gabriella Cagnoni
- Candiolo Cancer Institute - FPO (IRCCS) and University of Torino, Department of Oncology, Candiolo, Italy
| | - Sarah Gallet
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Naresh Kumar Hanchate
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Danièle Mazur
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
| | - Masahiko Taniguchi
- Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Massimiliano Mazzone
- Versalius Research Center, VIB, Laboratory of Molecular Oncology and Angiogenesis, Leuven, Belgium
- KU Keuven, Versalius Research Center, Leuven, Belgium
| | - Joost Verhaagen
- Netherlands institute for Neuroscience, Amsterdam, The Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philippe Ciofi
- INSERM, Neurocentre Magendie, U862, Université de Bordeaux, Bordeaux, France
| | - Sébastien G. Bouret
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
- The Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California, United States of America
| | - Luca Tamagnone
- Candiolo Cancer Institute - FPO (IRCCS) and University of Torino, Department of Oncology, Candiolo, Italy
| | - Vincent Prevot
- INSERM, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the Postnatal Brain, Lille, France
- UDSL, School of Medicine, Place de Verdun, Lille, France
- Institut de Médecine Prédictive et de Recherche Thérapeutique, IFR114, Lille, France
- * E-mail:
| |
Collapse
|
46
|
Li H, Yang X, Shi W, Ma Z, Feng G, Wang Q, Shen L, Xie C. Protective effects of nimodipine on cerebrovascular function in chronic alcoholic encephalopathy. Int J Mol Med 2013; 33:201-8. [PMID: 24173596 DOI: 10.3892/ijmm.2013.1540] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/17/2013] [Indexed: 11/05/2022] Open
Abstract
In the present study, we used chronic gavage administration of alcohol with gradual increases in alcohol concentration and volume to generate a rat model of chronic alcohol intoxication. We measured the changes in biological, behavioral, pathological and vascular injury-related molecular biological markers, and explored the effects of nimodipine intervention on alcohol intoxication. We found that chronic alcohol consumption induced a variety of behavioral abnormalities, accompanied by severe pathological changes in cerebral arterioles, prefrontal cortex and cerebellar tissue, as well as an upregulation of vascular endothelial growth factor (VEGF), leptin receptor (ob-R) and endothelin-1 (ET-1). Treatment with mimodipine for 15 days significantly improved behavioral abnormalities, alleviated the pathological changes in blood vessels and brain tissues, increased VEGF expression, decreased ob-R expression, reduced plasma ET-1 leakage and protected vascular and neuronal functions.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, Jinan Fourth People's Hospital, Jinan, Shandong, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Hao T, Rockwell P. Signaling through the vascular endothelial growth factor receptor VEGFR-2 protects hippocampal neurons from mitochondrial dysfunction and oxidative stress. Free Radic Biol Med 2013; 63:421-31. [PMID: 23732519 PMCID: PMC3756493 DOI: 10.1016/j.freeradbiomed.2013.05.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/28/2013] [Accepted: 05/24/2013] [Indexed: 12/12/2022]
Abstract
Vascular endothelial growth factor VEGF (VEGF-A or VEGF₁₆₅) is a potent angiogenic factor that also signals neuroprotection through activation of its cognate receptor VEGFR-2. In this capacity, VEGF signaling can rescue neurons from the damage induced by stressful stimuli many of which elicit oxidative stress. However, the regulatory role that VEGFR-2 plays in providing neuroprotection remains elusive. Therefore, we investigated the effects of VEGFR-2 inhibition on primary cultures of mature hippocampal neurons undergoing nutritional stress. We found that neurons cultured under nutritional stress had increased expression of VEGF and its receptors, VEGFR-1, VEGFR-2, and NP-1, as well as enhanced levels of VEGFR-2 phosphorylation. These neurons also showed increased activation of the prosurvival pathways for MEK/ERK1/2 and PI3K/Akt, enhanced phosphorylation (inactivation) of the proapoptotic BAD, and higher levels of the antiapoptotic protein Bcl-xL, all of which were augmented by treatments with exogenous VEGF and blocked by VEGFR-2 inhibition. The blockade of VEGFR-2 function also elicited a cytotoxicity that was accompanied by caspase-3 activation, induction of hemeoxygenase-1 (HO-1), oxidative stress, and a collapse in the mitochondrial membrane potential (ΔΨ(m)). Knockdown of VEGFR-2 by siRNA generated a similar pattern of redox change and mitochondrial impairment. Pretreatments with VEGF, VEGF-B, or the antioxidant N-acetylcysteine (NAC) rescued SU1498 or siRNA-treated neurons from the mitochondrial dysfunction and oxidative stress induced by VEGFR-2 inhibition in a timely fashion. These findings suggested that VEGF or VEGF-B can provide neuroprotection by signaling through an alternate VEGF receptor. Together, our findings suggest that VEGF signaling through VEGFR-2 plays a critical regulatory role in protecting stressed hippocampal neurons from the damaging effects of an oxidative insult. These findings also implicate VEGFR-1 or NP-1 as compensatory receptors that mediate neuroprotection when VEGFR-2 function is blocked.
Collapse
Affiliation(s)
- Tianfeng Hao
- Department of Biological Sciences, Hunter College and the Graduate Center of The City University of New York, 695 Park Ave, New York, NY 10065, USA
| | - Patricia Rockwell
- Department of Biological Sciences, Hunter College and the Graduate Center of The City University of New York, 695 Park Ave, New York, NY 10065, USA
| |
Collapse
|
48
|
Messina A, Giacobini P. Semaphorin signaling in the development and function of the gonadotropin hormone-releasing hormone system. Front Endocrinol (Lausanne) 2013; 4:133. [PMID: 24065959 PMCID: PMC3779810 DOI: 10.3389/fendo.2013.00133] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
The semaphorin proteins are among the best-studied families of guidance cues, contributing to morphogenesis and homeostasis in a wide range of tissue types. The major semaphorin receptors are plexins and neuropilins, however other receptors and co-receptors are capable to mediate signaling by semaphorins. These guidance proteins were originally identified as growth cone "collapsing factors" or as inhibitory signals, crucial for nervous system development. Since those seminal discoveries, the list of functions of semaphorins has rapidly grown. Over the past few years, a growing body of data indicates that semaphorins are involved in the regulation of the immune and vascular systems, in tumor growth/cancer cell metastasis and in neural circuit formation. Recently there has been increasing emphasis on research to determine the potential influence of semaphorins on the development and homeostasis of hormone systems and how circulating reproductive hormones regulate their expression and functions. Here, we focus on the emerging role of semaphorins in the development, differentiation and plasticity of unique neurons that secrete gonadotropin-releasing hormone (GnRH), which are essential for the acquisition and maintenance of reproductive competence in all vertebrates. Genetic evidence is also provided showing that insufficient semaphorin signaling contributes to some forms of reproductive disorders in humans, characterized by the reduction or failure of sexual competence. Finally, we will review some studies with the goal of highlighting how the expression of semaphorins and their receptors might be regulated by gonadal hormones in physiological and pathological conditions.
Collapse
Affiliation(s)
- Andrea Messina
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
| | - Paolo Giacobini
- INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unité 837, Lille, France
- School of Medicine, UDSL, Lille, France
- *Correspondence: Paolo Giacobini, INSERM, Laboratory of Development and Plasticity of the Postnatal Brain, Jean-Pierre Aubert Research Center, Unit 837, Place de Verdun, 59045 Lille Cedex, France e-mail:
| |
Collapse
|
49
|
Nivet AL, Vigneault C, Blondin P, Sirard MA. Changes in granulosa cells' gene expression associated with increased oocyte competence in bovine. Reproduction 2013; 145:555-65. [PMID: 23564726 DOI: 10.1530/rep-13-0032] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One of the challenges in mammalian reproduction is to understand the basic physiology of oocyte quality. It is believed that the follicle status is linked to developmental competence of the enclosed oocyte. To explore the link between follicles and competence in cows, previous research at our laboratory has developed an ovarian stimulation protocol that increases and then decreases oocyte quality according to the timing of oocyte recovery post-FSH withdrawal (coasting). Using this protocol, we have obtained the granulosa cells associated with oocytes of different qualities at selected times of coasting. Transcriptome analysis was done with Embryogene microarray slides and validation was performed by real-time PCR. Results show that the major changes in gene expression occurred from 20 to 44 h of coasting, when oocyte quality increases. Secondly, among upregulated genes (20-44 h), 25% were extracellular molecules, highlighting potential granulosa signaling cascades. Principal component analysis identified two patterns: one resembling the competence profile and another associated with follicle growth and atresia. Additionally, three major functional changes were identified: (i) the end of follicle growth (BMPR1B, IGF2, and RELN), involving interactions with the extracellular matrix (TFPI2); angiogenesis (NRP1), including early hypoxia, and potentially oxidative stress (GFPT2, TF, and VNN1) and (ii) apoptosis (KCNJ8) followed by iii) inflammation (ANKRD1). This unique window of analysis indicates a progressive hypoxia during coasting mixed with an increase in apoptosis and inflammation. Potential signaling pathways leading to competence have been identified and will require downstream testing. This preliminary analysis supports the potential role of the follicular differentiation in oocyte quality both during competence increase and decrease phases.
Collapse
Affiliation(s)
- Anne-Laure Nivet
- Département des sciences animales, Pavillon INAF, Faculté des sciences de l'agriculture et de l'alimentation, Centre de recherche en biologie de la reproduction, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | | | | | | |
Collapse
|
50
|
Chauvet S, Burk K, Mann F. Navigation rules for vessels and neurons: cooperative signaling between VEGF and neural guidance cues. Cell Mol Life Sci 2013; 70:1685-703. [PMID: 23475066 PMCID: PMC11113827 DOI: 10.1007/s00018-013-1278-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/22/2022]
Abstract
Many organs, such as lungs, nerves, blood and lymphatic vessels, consist of complex networks that carry flows of information, gases, and nutrients within the body. The morphogenetic patterning that generates these organs involves the coordinated action of developmental signaling cues that guide migration of specialized cells. Precision guidance of endothelial tip cells by vascular endothelial growth factors (VEGFs) is well established, and several families of neural guidance molecules have been identified to exert guidance function in both the nervous and the vascular systems. This review discusses recent advances in VEGF research, focusing on the emerging role of neural guidance molecules as key regulators of VEGF function during vascular development and on the novel role of VEGFs in neural cell migration and nerve wiring.
Collapse
Affiliation(s)
- Sophie Chauvet
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| | - Katja Burk
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| | - Fanny Mann
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Campus de Luminy Case 908, 13288 Marseille Cedex 9, France
| |
Collapse
|