1
|
Xuan F, Liu N, Zhang BX, Wen WX, Wang YC, Zhang HF, Wu XL. High expression and regulatory mechanisms of ANGPT1 and HOXA3 in acute myeloid leukemia. Bull Cancer 2025:S0007-4551(25)00118-3. [PMID: 40107921 DOI: 10.1016/j.bulcan.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 12/05/2024] [Accepted: 01/29/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Acute Myeloid Leukemia (AML) is a type of leukemia characterized by the malignant clonal proliferation of hematopoietic stem cells in the bone marrow. This study aims to investigate the role of ANGPT1 and HOXA3 in the leukemia cell line KG-1a. METHODS The expression patterns of ANGPT1 and HOXA3 in AML patients were determined by analyzing the TCGA database and clinical samples. Experiments were conducted using the KG-1a cell line, including flow cytometry and SA-β-Gal staining, to knock down ANGPT1 and HOXA3 and evaluate their functions. RESULTS ANGPT1 and HOXA3 were found to be highly expressed in AML patients. Knocking down ANGPT1 and HOXA3 promoted apoptosis and senescence in KG-1a cells by inhibiting proliferation-related genes and upregulating apoptosis-related genes. There is a reciprocal regulatory relationship between ANGPT1 and HOXA3, forming a positive feedback loop. Treatment with ATRA downregulated the expression of HOXA3 and induced apoptosis in KG-1a cells, highlighting the importance of HOXA3 as a therapeutic target in AML. CONCLUSION ANGPT1 and HOXA3 are highly expressed in AML, and knocking them down can promote apoptosis and senescence in leukemia cells. They exhibit a mutual regulatory relationship, forming a positive feedback loop. These findings contribute to a better understanding of the functional roles and regulatory mechanisms of ANGPT1 and HOXA3, and provide new scientific evidence for the treatment and prognosis improvement of AML patients.
Collapse
Affiliation(s)
- Fan Xuan
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Na Liu
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Bao-Xi Zhang
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Wen-Xiao Wen
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Yong-Cai Wang
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Hui-Feng Zhang
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Xiao-Li Wu
- Department of Pediatrics Hematology-Oncology, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
2
|
Fu Y, Zhang X, Wu H, Zhang P, Liu S, Guo T, Shan H, Liang Y, Chen H, Xie J, Duan Y. HOXA3 functions as the on-off switch to regulate the development of hESC-derived third pharyngeal pouch endoderm through EPHB2-mediated Wnt pathway. Front Immunol 2024; 14:1258074. [PMID: 38259452 PMCID: PMC10800530 DOI: 10.3389/fimmu.2023.1258074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
Objectives Normal commitment of the endoderm of the third pharyngeal pouch (3PP) is essential for the development and differentiation of the thymus. The aim of this study was to investigate the role of transcription factor HOXA3 in the development and differentiation of 3PP endoderm (3PPE) from human embryonic stem cells (hESCs). Methods The 3PPE was differentiated from hESC-derived definitive endoderm (DE) by mimicking developmental queues with Activin A, WNT3A, retinoic acid and BMP4. The function of 3PPE was assessed by further differentiating into functional thymic epithelial cells (TECs). The effect of HOXA3 inhibition on cells of 3PPE was subsequently investigated. Results A highly efficient approach for differentiating 3PPE cells was developed and these cells expressed 3PPE related genes HOXA3, SIX1, PAX9 as well as EpCAM. 3PPE cells had a strong potential to develop into TECs which expressed both cortical TEC markers K8 and CD205, and medullary TEC markers K5 and AIRE, and also promoted the development and maturation of T cells. More importantly, transcription factor HOXA3 not only regulated the differentiation of 3PPE, but also had a crucial role for the proliferation and migration of 3PPE cells. Our further investigation revealed that HOXA3 controlled the commitment and function of 3PPE through the regulation of Wnt signaling pathway by activating EPHB2. Conclusion Our results demonstrated that HOXA3 functioned as the on-off switch to regulate the development of hESC-derived 3PPE through EPHB2-mediated Wnt pathway, and our findings will provide new insights into studying the development of 3PP and thymic organ in vitro and in vivo.
Collapse
Affiliation(s)
- Yingjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Pingping Zhang
- Department of Laboratory Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Tingting Guo
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanhuan Shan
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yan Liang
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Clinical Medicine, the Second Affiliation Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
3
|
Kameda Y. Cellular and molecular mechanisms of the organogenesis and development, and function of the mammalian parathyroid gland. Cell Tissue Res 2023; 393:425-442. [PMID: 37410127 DOI: 10.1007/s00441-023-03785-3] [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: 03/18/2022] [Accepted: 05/12/2023] [Indexed: 07/07/2023]
Abstract
Serum calcium homeostasis is mainly regulated by parathormone (PTH) secreted by the parathyroid gland. Besides PTH and Gcm2, a master gene for parathyroid differentiation, many genes are expressed in the gland. Especially, calcium-sensing receptor (CaSR), vitamin D receptor (VDR), and Klotho function to prevent increased secretion of PTH and hyperplasia of the parathyroid gland under chronic hypocalcemia. Parathyroid-specific dual deletion of Klotho and CaSR induces a marked enlargement of the glandular size. The parathyroid develops from the third and fourth pharyngeal pouches except murine species in which the gland is derived from the third pouch only. The development of the murine parathyroid gland is categorized as follows: (1) formation and differentiation of the pharyngeal pouches, (2) appearance of parathyroid domain in the third pharyngeal pouch together with thymus domain, (3) migration of parathyroid primordium attached to the top of thymus, and (4) contact with the thyroid lobe and separation from the thymus. The transcription factors and signaling molecules involved in each of these developmental stages are elaborated. In addition, mesenchymal neural crest cells surrounding the pharyngeal pouches and parathyroid primordium and invading the parathyroid parenchyma participate in the development of the gland.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
4
|
Geggel RL, Prakash A, Emani SM, Sanders SP. Double Aortic Arch with Atresia of the Left Aortic Arch Proximal to the Left Common Carotid Artery: First Report of Documentation by Surgical Observation. Pediatr Cardiol 2023; 44:249-253. [PMID: 35930036 DOI: 10.1007/s00246-022-02979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 01/24/2023]
Abstract
Double aortic arch associated with atresia of the left arch proximal to the left common carotid artery has been considered a theoretical possibility. To our knowledge, we report the first patient with this anatomy confirmed by surgical observation.
Collapse
Affiliation(s)
- Robert L Geggel
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
| | - Ashwin Prakash
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Sitaram M Emani
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Stephen P Sanders
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| |
Collapse
|
5
|
Liu J, Zhang M, Dong H, Liu J, Mao A, Ning G, Cao Y, Zhang Y, Wang Q. Chemokine signaling synchronizes angioblast proliferation and differentiation during pharyngeal arch artery vasculogenesis. Development 2022; 149:285824. [PMID: 36468454 PMCID: PMC10114070 DOI: 10.1242/dev.200754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/14/2022] [Indexed: 12/09/2022]
Abstract
Developmentally, the great vessels of the heart originate from the pharyngeal arch arteries (PAAs). During PAA vasculogenesis, PAA precursors undergo sequential cell fate decisions that are accompanied by proliferative expansion. However, how these two processes are synchronized remains poorly understood. Here, we find that the zebrafish chemokine receptor Cxcr4a is expressed in PAA precursors, and genetic ablation of either cxcr4a or the ligand gene cxcl12b causes PAA stenosis. Cxcr4a is required for the activation of the downstream PI3K/AKT cascade, which promotes not only PAA angioblast proliferation, but also differentiation. AKT has a well-known role in accelerating cell-cycle progression through the activation of cyclin-dependent kinases. Despite this, we demonstrate that AKT phosphorylates Etv2 and Scl, the key regulators of angioblast commitment, on conserved serine residues, thereby protecting them from ubiquitin-mediated proteasomal degradation. Altogether, our study reveals a central role for chemokine signaling in PAA vasculogenesis through orchestrating angioblast proliferation and differentiation.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Haojian Dong
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jingwen Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
6
|
Zeng Z, Hu C, Ruan W, Zhang J, Lei S, Yang Y, Peng P, Pan F, Chen T. A specific immune signature for predicting the prognosis of glioma patients with IDH1-mutation and guiding immune checkpoint blockade therapy. Front Immunol 2022; 13:1001381. [PMID: 36159801 PMCID: PMC9500319 DOI: 10.3389/fimmu.2022.1001381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Isocitrate dehydrogenase (IDH1) is frequently mutated in glioma tissues, and this mutation mediates specific tumor-promoting mechanisms in glioma cells. We aimed to identify specific immune biomarkers for IDH1-mutation (IDH1mt) glioma. The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) were used to obtain RNA sequencing data and clinical characteristics of glioma tissues, while the stromal and immune scores of TCGA glioma tissues were determined using the ESTIMATE algorithm. Differentially expressed genes (DEGs), the protein–protein interaction(PPI) network, and least absolute shrinkage and selection operator (LASSO) and Cox regression analyses were used to select hub genes associated with stroma and immune scores and the prognoses of patients and to construct the risk model. The practicability and specificity of the risk model in both IDH1mt and IDH1-wildtype (wtIDH1) gliomas in TCGA and CGGA were evaluated. Molecular mechanisms, immunological characteristics and benefits of immune checkpoint blockade therapy in glioma tissues with IDH1mt were analyzed using GSEA, immunohistochemical staining, CIBERSORT, and T-cell dysfunction and exclusion (TIDE) analysis. The overall survival rate for IDH1mt-glioma patients with high stroma/immune scores was lower than that for those with low stroma/immune scores. A total of 222 DEGs were identified in IDH1mt glioma tissues with high stroma/immune scores. Among them, 72 genes had interactions in the PPI network, while three genes, HLA-DQA2, HOXA3, and SAA2, were selected as hub genes and used to construct risk models classifying patients into high- and low-risk score groups, followed by LASSO and Cox regression analyses. This risk model showed prognostic value in IDH1mt glioma in both TCGA and CCGA; nevertheless, the model was not suitable for wtIDH1 glioma. The risk model may act as an independent prognostic factor for IDH1mt glioma. IDH1mt glioma tissues from patients with high-risk scores showed more infiltration of M1 and CD8 T cells than those from patients with low-risk scores. Moreover, TIDE analysis showed that immune checkpoint blockade(ICB) therapy was highly beneficial for IDH1mt patients with high-risk scores. The risk model showed specific potential to predict the prognosis of IDH1mt-glioma patients, as well as guide ICB, contributing to the diagnosis and therapy of IDH1mt-glioma patients.
Collapse
Affiliation(s)
- Zhirui Zeng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Chujiao Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Wanyuan Ruan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jinjuan Zhang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Shan Lei
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Yushi Yang
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Pailan Peng
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- *Correspondence: Pailan Peng, ; Feng Pan, ; Tengxiang Chen,
| | - Feng Pan
- Department of Bone and Joint Surgery, Gui Zhou Orthopedic Hospital, Guiyang, China
- *Correspondence: Pailan Peng, ; Feng Pan, ; Tengxiang Chen,
| | - Tengxiang Chen
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
- *Correspondence: Pailan Peng, ; Feng Pan, ; Tengxiang Chen,
| |
Collapse
|
7
|
Anderson RH, Bamforth SD. Morphogenesis of the Mammalian Aortic Arch Arteries. Front Cell Dev Biol 2022; 10:892900. [PMID: 35620058 PMCID: PMC9127140 DOI: 10.3389/fcell.2022.892900] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/20/2022] [Indexed: 12/02/2022] Open
Abstract
The major vessels in mammals that take blood away from the heart and deliver it to the arms and the head take their origin from the aortic arch and are derived from the arteries formed within the embryonic pharyngeal arches. These pharyngeal arch arteries, initially symmetrical, form in a cranial to caudal sequence within the pharyngeal mesenchyme. They then undergo a complex process of remodeling to produce the asymmetrical brachiocephalic arteries as seen in the adult. A complex interaction between the tissues of the pharyngeal arches and the genes they express is required to ensure that arterial formation and remodeling is able to proceed normally. If this process is disrupted, life-threatening congenital cardiovascular malformations can occur, such as interruption of the aortic arch, isolation of individual arteries, or so-called vascular rings. Here, using state-of-the-art imaging techniques, we describe the morphogenesis of the arteries in humans and mice and the cardiovascular defects in the Tbx1 mutant mouse model. We provide details of the process of remodeling, clarifying also the morphogenesis of the external carotid artery and the so-called "migration" of the left subclavian artery.
Collapse
|
8
|
Pleș L, Cîrstoveanu C, Sima RM, Gorecki GP, Chicea R, Haj Hamoud B. Prenatal Diagnosis of Bovine Aortic Arch Anatomic Variant. Diagnostics (Basel) 2022; 12:diagnostics12030624. [PMID: 35328177 PMCID: PMC8947431 DOI: 10.3390/diagnostics12030624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 11/16/2022] Open
Abstract
Fetal aortic arch development is an early and complex process that depends on many genetic and environmental factors. The final aortic arch varies greatly; it may take the form of a normal arch, anatomic variant (AAAV) with a common origin to that of the innominate artery and left common carotid artery (formerly known as “bovine aortic arch” (with an incidence of up to 27%)) or one of multiple pathological conditions. The present study aimed to establish the feasibility and impact of prenatal anatomic arch variants’ diagnosis. A retrospective study of 271 fetal second- and third-trimester anomaly scans was performed in our tertiary center. Examinations that evaluated the sagittal aortic arch were included and the branching pattern was assessed. Additionally, a literature data search based on the terms “common origin of innominate artery and left common carotid artery”, “bovine arch”, “bovine aortic” and “aortic arch anomalies” was performed. Results that referred to prenatal AAAV were retained and the papers evaluated. In our study, the AAA incidence was 1.93%, with 4 out of 5 cases being arch type B. All cases had minor associated conditions but a good postnatal outcome. An anatomic aortic variant with a common IA and LCCa prenatal diagnosis was found in a small number of studies; most of the cases described in pediatric and adult series were related to cardiac surgery for stenting, aneurysm or thoracic-associated diseases. The incidence of AAAV varied from 6 to 27% depending on the population studied (highest incidence in African individuals). The variant was highly associated with aortic dissection, pulmonary and cerebral embolism and increased risks of incidents during surgery. Diagnosing AAAV during a routine anatomic scan is feasible and diagnoses can be made when anomaly scans are performed. Awareness of the condition is important for postnatal surgery when other cardiac anomalies are found; this can prevent accidents with simple changes to the patient’s lifestyle, and, in the case of surgery, means we can adopt the correct surgical approach.
Collapse
Affiliation(s)
- Liana Pleș
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- The “Bucur” Maternity, ‘Saint John’ Hospital, 040294 Bucharest, Romania
| | - Cătălin Cîrstoveanu
- Department of Pediatrics, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Pediatrics Department, ‘Maria Sklodowska Curie’ Emergency Children Clinical Hospital, 041451 Bucharest, Romania
| | - Romina-Marina Sima
- Department of Obstetrics and Gynecology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- The “Bucur” Maternity, ‘Saint John’ Hospital, 040294 Bucharest, Romania
- Correspondence: (R.-M.S.); (G.-P.G.)
| | - Gabriel-Petre Gorecki
- The “Bucur” Maternity, ‘Saint John’ Hospital, 040294 Bucharest, Romania
- Faculty of Medicine, ‘Titu Maiorescu’ University, 040441 Bucharest, Romania
- Correspondence: (R.-M.S.); (G.-P.G.)
| | - Radu Chicea
- Faculty of Medicine, ‘Lucian Blaga’ University of Sibiu, 550024 Sibiu, Romania;
| | - Bashar Haj Hamoud
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Hospital, Kirrberger Straße 100, Building 9, 66421 Homburg, Germany;
| |
Collapse
|
9
|
Ye S, Yang N, Lu T, Wu T, Wang L, Pan YH, Cao X, Yuan X, Wisniewski T, Dang S, Zhang W. Adamts18 modulates the development of the aortic arch and common carotid artery. iScience 2021; 24:102672. [PMID: 34189436 PMCID: PMC8215225 DOI: 10.1016/j.isci.2021.102672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023] Open
Abstract
Members of a disintegrin and metalloproteinases with thrombospondin motif (ADAMTS) family have been implicated in various vascular diseases. However, their functional roles in early embryonic vascular development are unknown. In this study, we showed that Adamts18 is highly expressed at E11.5-E14.5 in cells surrounding the embryonic aortic arch (AOAR) and the common carotid artery (CCA) during branchial arch artery development in mice. Adamts18 deficiency was found to cause abnormal development of AOAR, CCA, and the third and fourth branchial arch appendages, leading to hypoplastic carotid body, thymus, and variation of middle cerebral artery. Adamts18 was shown to affect the accumulation of extracellular matrix (ECM) components, in particular fibronectin (Fn), around AOAR and CCA. As a result of increased Fn accumulation, the Notch3 signaling pathway was activated to promote the differentiation of cranial neural crest cells (CNCCs) to vascular smooth muscle cells. These data indicate that Adamts18-mediated ECM homeostasis is crucial for the differentiation of CNCCs.
Collapse
Affiliation(s)
- Shuai Ye
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Ning Yang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Tiantian Lu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Taojing Wu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Liya Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Xiaohua Cao
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Xiaobing Yuan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, New York University Langone Health, New York, NY, USA
| | - Suying Dang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
10
|
Guida V, Calzari L, Fadda MT, Piceci-Sparascio F, Digilio MC, Bernardini L, Brancati F, Mattina T, Melis D, Forzano F, Briuglia S, Mazza T, Bianca S, Valente EM, Salehi LB, Prontera P, Pagnoni M, Tenconi R, Dallapiccola B, Iannetti G, Corsaro L, De Luca A, Gentilini D. Genome-Wide DNA Methylation Analysis of a Cohort of 41 Patients Affected by Oculo-Auriculo-Vertebral Spectrum (OAVS). Int J Mol Sci 2021; 22:ijms22031190. [PMID: 33530447 PMCID: PMC7866060 DOI: 10.3390/ijms22031190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Oculo-auriculo-vertebral-spectrum (OAVS; OMIM 164210) is a rare disorder originating from abnormal development of the first and second branchial arch. The clinical phenotype is extremely heterogeneous with ear anomalies, hemifacial microsomia, ocular defects, and vertebral malformations being the main features. MYT1, AMIGO2, and ZYG11B gene variants were reported in a few OAVS patients, but the etiology remains largely unknown. A multifactorial origin has been proposed, including the involvement of environmental and epigenetic mechanisms. To identify the epigenetic mechanisms contributing to OAVS, we evaluated the DNA-methylation profiles of 41 OAVS unrelated affected individuals by using a genome-wide microarray-based methylation approach. The analysis was first carried out comparing OAVS patients with controls at the group level. It revealed a moderate epigenetic variation in a large number of genes implicated in basic chromatin dynamics such as DNA packaging and protein-DNA organization. The alternative analysis in individual profiles based on the searching for Stochastic Epigenetic Variants (SEV) identified an increased number of SEVs in OAVS patients compared to controls. Although no recurrent deregulated enriched regions were found, isolated patients harboring suggestive epigenetic deregulations were identified. The recognition of a different DNA methylation pattern in the OAVS cohort and the identification of isolated patients with suggestive epigenetic variations provide consistent evidence for the contribution of epigenetic mechanisms to the etiology of this complex and heterogeneous disorder.
Collapse
Affiliation(s)
- Valentina Guida
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.P.-S.); (L.B.); (A.D.L.)
- Correspondence: (V.G.); (D.G.)
| | - Luciano Calzari
- Istituto Auxologico Italiano IRCCS, Bioinformatics and Statistical Genomics Unit, Cusano Milanino, 20095 Milano, Italy;
| | - Maria Teresa Fadda
- Department of Maxillofacial Surgery, Sapienza University of Rome, 00161 Rome, Italy; (M.T.F.); (M.P.); (G.I.)
| | - Francesca Piceci-Sparascio
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.P.-S.); (L.B.); (A.D.L.)
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy; (M.C.D.); (B.D.)
| | - Laura Bernardini
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.P.-S.); (L.B.); (A.D.L.)
| | - Francesco Brancati
- Department of Life, Health and Environmental Sciences, Unit of Medical Genetics University of L’Aquila, 67100 L’Aquila, Italy;
- IRCCS San Raffaele Pisana, 00163 Rome, Italy
| | - Teresa Mattina
- Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy;
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Salerno, Italy;
| | - Francesca Forzano
- Clinical Genetics Department, Guy’s & St Thomas’ NHS Foundation Trust, London SE1 7EH, UK;
| | | | - Tommaso Mazza
- Unit of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy;
| | - Sebastiano Bianca
- Centro di Consulenza Genetica e Teratologia della Riproduzione, Dipartimento Materno Infantile, ARNAS Garibaldi Nesima, 95123 Catania, Italy;
| | - Enza Maria Valente
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | | | - Paolo Prontera
- Medical Genetics Unit, University of Perugia Hospital SM della Misericordia, 06129 Perugia, Italy;
| | - Mario Pagnoni
- Department of Maxillofacial Surgery, Sapienza University of Rome, 00161 Rome, Italy; (M.T.F.); (M.P.); (G.I.)
| | - Romano Tenconi
- Department of Pediatrics, Clinical Genetics, Università di Padova, 35122 Padova, Italy;
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy; (M.C.D.); (B.D.)
| | - Giorgio Iannetti
- Department of Maxillofacial Surgery, Sapienza University of Rome, 00161 Rome, Italy; (M.T.F.); (M.P.); (G.I.)
| | - Luigi Corsaro
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Alessandro De Luca
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (F.P.-S.); (L.B.); (A.D.L.)
| | - Davide Gentilini
- Istituto Auxologico Italiano IRCCS, Bioinformatics and Statistical Genomics Unit, Cusano Milanino, 20095 Milano, Italy;
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- Correspondence: (V.G.); (D.G.)
| |
Collapse
|
11
|
Mao A, Zhang M, Li L, Liu J, Ning G, Cao Y, Wang Q. Pharyngeal pouches provide a niche microenvironment for arch artery progenitor specification. Development 2021; 148:dev.192658. [PMID: 33334861 PMCID: PMC7847271 DOI: 10.1242/dev.192658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/10/2020] [Indexed: 11/20/2022]
Abstract
The paired pharyngeal arch arteries (PAAs) are transient blood vessels connecting the heart with the dorsal aorta during embryogenesis. Although PAA malformations often occur along with pharyngeal pouch defects, the functional interaction between these adjacent tissues remains largely unclear. Here, we report that pharyngeal pouches are essential for PAA progenitor specification in zebrafish embryos. We reveal that the segmentation of pharyngeal pouches coincides spatiotemporally with the emergence of PAA progenitor clusters. These pouches physically associate with pharyngeal mesoderm in discrete regions and provide a niche microenvironment for PAA progenitor commitment by expressing BMP proteins. Specifically, pouch-derived BMP2a and BMP5 are the primary niche cues responsible for activating the BMP/Smad pathway in pharyngeal mesoderm, thereby promoting progenitor specification. In addition, BMP2a and BMP5 play an inductive function in the expression of the cloche gene npas4l in PAA progenitors. cloche mutants exhibit a striking failure to specify PAA progenitors and display ectopic expression of head muscle markers in the pharyngeal mesoderm. Therefore, our results support a crucial role for pharyngeal pouches in establishing a progenitor niche for PAA morphogenesis via BMP2a/5 expression.
Collapse
Affiliation(s)
- Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Linwei Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
12
|
Kameda Y. Molecular and cellular mechanisms of the organogenesis and development of the mammalian carotid body. Dev Dyn 2019; 249:592-609. [DOI: 10.1002/dvdy.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/08/2019] [Accepted: 12/08/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
- Yoko Kameda
- Department of AnatomyKitasato University School of Medicine Sagamihara Japan
| |
Collapse
|
13
|
Phillips HM, Stothard CA, Shaikh Qureshi WM, Kousa AI, Briones-Leon JA, Khasawneh RR, O'Loughlin C, Sanders R, Mazzotta S, Dodds R, Seidel K, Bates T, Nakatomi M, Cockell SJ, Schneider JE, Mohun TJ, Maehr R, Kist R, Peters H, Bamforth SD. Pax9 is required for cardiovascular development and interacts with Tbx1 in the pharyngeal endoderm to control 4th pharyngeal arch artery morphogenesis. Development 2019; 146:dev.177618. [PMID: 31444215 PMCID: PMC6765178 DOI: 10.1242/dev.177618] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022]
Abstract
Developmental defects affecting the heart and aortic arch arteries are a significant phenotype observed in individuals with 22q11 deletion syndrome and are caused by a microdeletion on chromosome 22q11. TBX1, one of the deleted genes, is expressed throughout the pharyngeal arches and is considered a key gene, when mutated, for the arch artery defects. Pax9 is expressed in the pharyngeal endoderm and is downregulated in Tbx1 mutant mice. We show here that Pax9-deficient mice are born with complex cardiovascular malformations that affect the outflow tract and aortic arch arteries with failure of the 3rd and 4th pharyngeal arch arteries to form correctly. Transcriptome analysis indicated that Pax9 and Tbx1 may function together, and mice double heterozygous for Tbx1/Pax9 presented with a significantly increased incidence of interrupted aortic arch when compared with Tbx1 heterozygous mice. Using a novel Pax9Cre allele, we demonstrated that the site of this Tbx1-Pax9 genetic interaction is the pharyngeal endoderm, therefore revealing that a Tbx1-Pax9-controlled signalling mechanism emanating from the pharyngeal endoderm is required for crucial tissue interactions during normal morphogenesis of the pharyngeal arch artery system. Summary: A strong genetic interaction between Tbx1 and Pax9 that leads to 4th PAA-derived defects in double heterozygous mice is cell-autonomous within the pharyngeal endoderm.
Collapse
Affiliation(s)
- Helen M Phillips
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Catherine A Stothard
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | | | | | | | - Ramada R Khasawneh
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Chloe O'Loughlin
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Rachel Sanders
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Silvia Mazzotta
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Rebecca Dodds
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Kerstin Seidel
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Timothy Bates
- School of Dental Sciences, Newcastle University, Newcastle-upon-Tyne NE2 4BW, UK
| | - Mitsushiro Nakatomi
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Simon J Cockell
- Bioinformatics Support Unit, Newcastle University, Newcastle-upon-Tyne NE2 4HH, UK
| | | | | | - René Maehr
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ralf Kist
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK.,School of Dental Sciences, Newcastle University, Newcastle-upon-Tyne NE2 4BW, UK
| | - Heiko Peters
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| | - Simon D Bamforth
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, UK
| |
Collapse
|
14
|
Sugrue KF, Zohn IE. Reduced maternal vitamin A status increases the incidence of normal aortic arch variants. Genesis 2019; 57:e23326. [PMID: 31299141 DOI: 10.1002/dvg.23326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022]
Abstract
While common in the general population, the developmental origins of "normal" anatomic variants of the aortic arch remain unknown. Aortic arch development begins with the establishment of the second heart field (SHF) that contributes to the pharyngeal arch arteries (PAAs). The PAAs remodel during subsequent development to form the mature aortic arch and arch vessels. Retinoic acid signaling involving the biologically active metabolite of vitamin A, plays a key role in multiple steps of this process. Recent work from our laboratory indicates that the E3 ubiquitin ligase Hectd1 is required for full activation of retinoic acid signaling during cardiac development. Furthermore, our study suggested that mild alterations in retinoic acid signaling combined with reduced gene dosage of Hectd1, results in a benign aortic arch variant where the transverse aortic arch is shortened between the brachiocephalic and left common carotid arteries. These abnormalities are preceded by hypoplasia of the fourth PAA. To further explore this interaction, we investigate whether reduced maternal dietary vitamin A intake can similarly influence aortic arch development. Our findings indicate that the incidence of hypoplastic fourth PAAs, as well as the incidence of shortened transverse arch are increased with reduced maternal vitamin A intake during pregnancy. These studies provide new insights as to the developmental origins of these benign aortic arch variants.
Collapse
Affiliation(s)
- Kelsey F Sugrue
- Institute for Biomedical Sciences, The George Washington University, Washington, District of Columbia.,Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia.,Center for Genetic Medicine, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia.,Center for Genetic Medicine, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia
| |
Collapse
|
15
|
Asymmetry in Mechanosensitive Gene Expression during Aortic Arch Morphogenesis. Sci Rep 2018; 8:16948. [PMID: 30446764 PMCID: PMC6240117 DOI: 10.1038/s41598-018-35127-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/30/2018] [Indexed: 11/09/2022] Open
Abstract
Embryonic aortic arches (AA) are initially bilaterally paired, transitional vessels and failures in remodeling based on hemodynamic and growth-related adaptations cause a spectrum of congenital heart disease (CHD) anatomies. Identifying regulatory mechanisms and cross-talk between the genetic elements of these vessels are critical to understand the ethiology of CHD and refine predictive computational models. This study aims to screen expression profiles of fundamental biological pathways in AA at early stages of chick embryo morphogenesis and correlate them with our current understanding of growth and mechanical loading. Reverse transcription-quantitative PCR (RT-qPCR) was followed by correlation and novel peak expression analyses to compare the behaviour and activation period of the genes. Available protein networks were also integrated to investigate the interactions between molecules and highlight major hierarchies. Only wall shear stress (WSS) and growth-correlated expression patterns were investigated. Effect of WSS was seen directly on angiogenesis as well on structural and apoptosis-related genes. Our time-resolved network suggested that WSS-correlated genes coordinate the activity of critical growth factors. Moreover, differential gene expression of left and right AA might be an indicator of subsequent asymmetric morphogenesis. These findings may further our understanding of the complex processes of cardiac morphogenesis and errors resulting in CHD.
Collapse
|
16
|
Abrial M, Paffett-Lugassy N, Jeffrey S, Jordan D, O'Loughlin E, Frederick CJ, Burns CG, Burns CE. TGF-β Signaling Is Necessary and Sufficient for Pharyngeal Arch Artery Angioblast Formation. Cell Rep 2018; 20:973-983. [PMID: 28746880 PMCID: PMC5565225 DOI: 10.1016/j.celrep.2017.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 05/23/2017] [Accepted: 06/30/2017] [Indexed: 11/15/2022] Open
Abstract
The pharyngeal arch arteries (PAAs) are transient embryonic blood vessels that mature into critical segments of the aortic arch and its branches. Although defects in PAA development cause life-threating congenital cardiovascular defects, the molecular mechanisms that orchestrate PAA morphogenesis remain unclear. Through small-molecule screening in zebrafish, we identified TGF-β signaling as indispensable for PAA development. Specifically, chemical inhibition of the TGF-β type I receptor ALK5 impairs PAA development because nkx2.5+ PAA progenitor cells fail to differentiate into tie1+ angioblasts. Consistent with this observation, we documented a burst of ALK5-mediated Smad3 phosphorylation within PAA progenitors that foreshadows angioblast emergence. Remarkably, premature induction of TGF-β receptor activity stimulates precocious angioblast differentiation, thereby demonstrating the sufficiency of this pathway for initiating the PAA progenitor to angioblast transition. More broadly, these data uncover TGF-β as a rare signaling pathway that is necessary and sufficient for angioblast lineage commitment.
Collapse
Affiliation(s)
- Maryline Abrial
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Noëlle Paffett-Lugassy
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Spencer Jeffrey
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Daniel Jordan
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Evan O'Loughlin
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Charles J Frederick
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - C Geoffrey Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Caroline E Burns
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
17
|
Losa M, Latorre V, Andrabi M, Ladam F, Sagerström C, Novoa A, Zarrineh P, Bridoux L, Hanley NA, Mallo M, Bobola N. A tissue-specific, Gata6-driven transcriptional program instructs remodeling of the mature arterial tree. eLife 2017; 6:31362. [PMID: 28952437 PMCID: PMC5630260 DOI: 10.7554/elife.31362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/25/2017] [Indexed: 01/23/2023] Open
Abstract
Connection of the heart to the systemic circulation is a critical developmental event that requires selective preservation of embryonic vessels (aortic arches). However, why some aortic arches regress while others are incorporated into the mature aortic tree remains unclear. By microdissection and deep sequencing in mouse, we find that neural crest (NC) only differentiates into vascular smooth muscle cells (SMCs) around those aortic arches destined for survival and reorganization, and identify the transcription factor Gata6 as a crucial regulator of this process. Gata6 is expressed in SMCs and its target genes activation control SMC differentiation. Furthermore, Gata6 is sufficient to promote SMCs differentiation in vivo, and drive preservation of aortic arches that ought to regress. These findings identify Gata6-directed differentiation of NC to SMCs as an essential mechanism that specifies the aortic tree, and provide a new framework for how mutations in GATA6 lead to congenital heart disorders in humans.
Collapse
Affiliation(s)
- Marta Losa
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Victor Latorre
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Munazah Andrabi
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Franck Ladam
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Charles Sagerström
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Ana Novoa
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Peyman Zarrineh
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Laure Bridoux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Neil A Hanley
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Endocrinology Department, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Moises Mallo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Nicoletta Bobola
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
18
|
Raimondi F, Bonnet D, Geva T, Sanders SP. Anomalous origin of the left innominate (brachiocephalic) artery in the right aortic arch: How can it be anomalous when the left innominate artery is absent? Ann Pediatr Cardiol 2016; 9:170-2. [PMID: 27212855 PMCID: PMC4867805 DOI: 10.4103/0974-2069.181497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
An unusual case of a rare vascular ring, which has been called right aortic arch with aberrant left innominate artery, is presented. The appearance of this case led to the realization that there is really no innominate artery present in this anomaly but only the left dorsal aorta. We present a clarification of the nature and likely development of the vessels present.
Collapse
Affiliation(s)
- Francesca Raimondi
- Centre de Référence Malformations Cardiaques Congénitales Complexes-M3C Université Paris Descartes, Sorbonne Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Damien Bonnet
- Centre de Référence Malformations Cardiaques Congénitales Complexes-M3C Université Paris Descartes, Sorbonne Paris Cité, Hôpital Necker-Enfants Malades, Paris, France
| | - Tal Geva
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen P Sanders
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, Pathology, and Cardiac Surgery, Boston Children's Hospital, Massachusetts, USA
| |
Collapse
|
19
|
Prakash S, Kuang SQ, Regalado E, Guo D, Milewicz D. Recurrent Rare Genomic Copy Number Variants and Bicuspid Aortic Valve Are Enriched in Early Onset Thoracic Aortic Aneurysms and Dissections. PLoS One 2016; 11:e0153543. [PMID: 27092555 PMCID: PMC4836726 DOI: 10.1371/journal.pone.0153543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 03/18/2016] [Indexed: 11/23/2022] Open
Abstract
Thoracic Aortic Aneurysms and Dissections (TAAD) are a major cause of death in the United States. The spectrum of TAAD ranges from genetic disorders, such as Marfan syndrome, to sporadic isolated disease of unknown cause. We hypothesized that genomic copy number variants (CNVs) contribute causally to early onset TAAD (ETAAD). We conducted a genome-wide SNP array analysis of ETAAD patients of European descent who were enrolled in the National Registry of Genetically Triggered Thoracic Aortic Aneurysms and Cardiovascular Conditions (GenTAC). Genotyping was performed on the Illumina Omni-Express platform, using PennCNV, Nexus and CNVPartition for CNV detection. ETAAD patients (n = 108, 100% European American, 28% female, average age 20 years, 55% with bicuspid aortic valves) were compared to 7013 dbGAP controls without a history of vascular disease using downsampled Omni 2.5 data. For comparison, 805 sporadic TAAD patients with late onset aortic disease (STAAD cohort) and 192 affected probands from families with at least two affected relatives (FTAAD cohort) from our institution were screened for additional CNVs at these loci with SNP arrays. We identified 47 recurrent CNV regions in the ETAAD, FTAAD and STAAD groups that were absent or extremely rare in controls. Nine rare CNVs that were either very large (>1 Mb) or shared by ETAAD and STAAD or FTAAD patients were also identified. Four rare CNVs involved genes that cause arterial aneurysms when mutated. The largest and most prevalent of the recurrent CNVs were at Xq28 (two duplications and two deletions) and 17q25.1 (three duplications). The percentage of individuals harboring rare CNVs was significantly greater in the ETAAD cohort (32%) than in the FTAAD (23%) or STAAD (17%) cohorts. We identified multiple loci affected by rare CNVs in one-third of ETAAD patients, confirming the genetic heterogeneity of TAAD. Alterations of candidate genes at these loci may contribute to the pathogenesis of TAAD.
Collapse
Affiliation(s)
- Siddharth Prakash
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Shao-Qing Kuang
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - GenTAC Registry Investigators
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Ellen Regalado
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dongchuan Guo
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dianna Milewicz
- Department of Internal Medicine, Division of Medical Genetics, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
20
|
Kameda Y. Cellular and molecular events on the development of mammalian thyroid C cells. Dev Dyn 2016; 245:323-41. [PMID: 26661795 DOI: 10.1002/dvdy.24377] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/05/2015] [Indexed: 12/12/2022] Open
Abstract
Thyroid C cells synthesize and secrete calcitonin, a serum calcium-lowering hormone. This review provides our current understanding of mammalian thyroid C cells from the molecular and morphological perspectives. Several transcription factors and signaling molecules involved in the development of C cells have been identified, and genes expressed in the pharyngeal pouch endoderm, neural crest-derived mesenchyme in the pharyngeal arches, and ultimobranchial body play critical roles for the development of C cells. It has been generally accepted, without much-supporting evidence, that mammalian C cells, as well as the avian cells, are derived from the neural crest. However, by fate mapping of neural crest cells in both Wnt1-Cre/R26R and Connexin(Cxn)43-lacZ transgenic mice, we showed that neural crest cells colonize neither the fourth pharyngeal pouch nor the ultimobranchial body. E-cadherin, an epithelial cell marker, is expressed in thyroid C cells and their precursors, the fourth pharyngeal pouch and ultimobranchial body. Furthermore, E-cadherin is colocalized with calcitonin in C cells. Recently, lineage tracing in Sox17-2A-iCre/R26R mice has clarified that the pharyngeal endoderm-derived cells give rise to C cells. Together, these findings indicate that mouse thyroid C cells are endodermal in origin.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| |
Collapse
|
21
|
Sherif HM. Heterogeneity in the Segmental Development of the Aortic Tree: Impact on Management of Genetically Triggered Aortic Aneurysms. AORTA (STAMFORD, CONN.) 2014; 2:186-95. [PMID: 26798739 PMCID: PMC4686358 DOI: 10.12945/j.aorta.2014.14-032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/07/2014] [Indexed: 11/18/2022]
Abstract
An extensive search of the medical literature examining the development of the thoracic aortic tree reveals that the thoracic aorta does not develop as one unit or in one stage: the oldest part of the thoracic aorta is the descending aorta with the aortic arch being the second oldest, developing under influence from the neural crest cell. Following in chronological order are the proximal ascending aorta and aortic root, which develop from a conotruncal origin. Different areas of the thoracic aorta develop under the influence of different gene sets. These parts develop from different cell lineages: the aortic root (the conotruncus), developing from the mesoderm; the ascending aorta and aortic arch, developing from the neural crest cells; and the descending aorta from the mesoderm. Findings illustrate that the thoracic aorta is not a single entity, in developmental terms. It develops from three or four distinct areas, at different stages of embryonic life, and under different sets of genes and signaling pathways. Genetically triggered thoracic aortic aneurysms are not a monolithic group but rather share a multi-genetic origin. Identification of therapeutic targets should be based on the predilection of certain genes to cause aneurysmal disease in specific aortic segments.
Collapse
Affiliation(s)
- Hisham M.F. Sherif
- Department of Cardiac Surgery, Christiana Hospital, Christiana Care Health System, Newark, Delaware, USA
| |
Collapse
|
22
|
Frisdal A, Trainor PA. Development and evolution of the pharyngeal apparatus. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:403-18. [PMID: 25176500 DOI: 10.1002/wdev.147] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/19/2014] [Accepted: 06/10/2014] [Indexed: 01/06/2023]
Abstract
The oral or pharyngeal apparatus facilitates the dual functions of respiration and feeding. It develops during embryogenesis from transient structures called pharyngeal arches (PAs), which comprise a reiterated series of outgrowths on the lateral side of the head. The PAs and their segmental arrangement are highly conserved throughout evolution from invertebrate chordates such as amphioxus, through to vertebrate agnathans including avians, squamates, and mammals. The structural organization of the PAs is also highly conserved and involves contributions from each of the three primary endoderm, mesoderm, and ectoderm germ layers. The endoderm is particularly important for PA formation and segmentation and also plays a critical role in tissue-specific differentiation. The ectoderm gives rise to neural crest cells (NCC) which provide an additional layer of complexity to PA development and differentiation in vertebrates compared to invertebrate chordates that do not possess NCC. Collectively, the PAs give rise to much of the neurovasculature and musculoskeletal systems in the head and neck. The complexity of development renders the pharyngeal apparatus prone to perturbation and subsequently the pathogenesis of birth defects. Hence it is important to understand the signals and mechanisms that govern the development and evolution of the pharyngeal complex.
Collapse
Affiliation(s)
- Aude Frisdal
- Stowers Institute for Medical Research, Kansas City, MO, USA; University Pierre and Marie Curie, Paris, France
| | | |
Collapse
|
23
|
Gonzalez C, Conde SV, Gallego-Martín T, Olea E, Gonzalez-Obeso E, Ramirez M, Yubero S, Agapito MT, Gomez-Niñno A, Obeso A, Rigual R, Rocher A. Fernando de Castro and the discovery of the arterial chemoreceptors. Front Neuroanat 2014; 8:25. [PMID: 24860435 PMCID: PMC4026738 DOI: 10.3389/fnana.2014.00025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/03/2014] [Indexed: 01/27/2023] Open
Abstract
When de Castro entered the carotid body (CB) field, the organ was considered to be a small autonomic ganglion, a gland, a glomus or glomerulus, or a paraganglion. In his 1928 paper, de Castro concluded: “In sum, the Glomus caroticum is innervated by centripetal fibers, whose trophic centers are located in the sensory ganglia of the glossopharyngeal, and not by centrifugal [efferent] or secretomotor fibers as is the case for glands; these are precisely the facts which lead to suppose that the Glomus caroticum is a sensory organ.” A few pages down, de Castro wrote: “The Glomus represents an organ with multiple receptors furnished with specialized receptor cells like those of other sensory organs [taste buds?]…As a plausible hypothesis we propose that the Glomus caroticum represents a sensory organ, at present the only one in its kind, dedicated to capture certain qualitative variations in the composition of blood, a function that, possibly by a reflex mechanism would have an effect on the functional activity of other organs… Therefore, the sensory fiber would not be directly stimulated by blood, but via the intermediation of the epithelial cells of the organ, which, as their structure suggests, possess a secretory function which would participate in the stimulation of the centripetal fibers.” In our article we will recreate the experiments that allowed Fernando de Castro to reach this first conclusion. Also, we will scrutinize the natural endowments and the scientific knowledge that drove de Castro to make the triple hypotheses: the CB as chemoreceptor (variations in blood composition), as a secondary sensory receptor which functioning involves a chemical synapse, and as a center, origin of systemic reflexes. After a brief account of the systemic reflex effects resulting from the CB stimulation, we will complete our article with a general view of the cellular-molecular mechanisms currently thought to be involved in the functioning of this arterial chemoreceptor.
Collapse
Affiliation(s)
- Constancio Gonzalez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Silvia V Conde
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Teresa Gallego-Martín
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Elena Olea
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Elvira Gonzalez-Obeso
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Maria Ramirez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Sara Yubero
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Maria T Agapito
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Angela Gomez-Niñno
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Ana Obeso
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Ricardo Rigual
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| | - Asunción Rocher
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas, Universidad de Valladolid Valladolid, España ; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Facultad de Medicina, Universidad de Valladolid Valladolid, España
| |
Collapse
|
24
|
Kameda Y. Signaling molecules and transcription factors involved in the development of the sympathetic nervous system, with special emphasis on the superior cervical ganglion. Cell Tissue Res 2014; 357:527-48. [PMID: 24770894 DOI: 10.1007/s00441-014-1847-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/12/2014] [Indexed: 12/16/2022]
Abstract
The cells that constitute the sympathetic nervous system originate from the neural crest. This review addresses the current understanding of sympathetic ganglion development viewed from molecular and morphological perspectives. Development of the sympathetic nervous system is categorized into three main steps, as follows: (1) differentiation and migration of cells in the neural crest lineage for formation of the primary sympathetic chain, (2) differentiation of sympathetic progenitors, and (3) growth and survival of sympathetic ganglia. The signaling molecules and transcription factors involved in each of these developmental stages are elaborated mostly on the basis of the results of targeted mutation of respective genes. Analyses in mutant mice revealed differences between the superior cervical ganglion (SCG) and the other posterior sympathetic ganglia. This review provides a summary of the similarities and differences in the development of the SCG and other posterior sympathetic ganglia. Relevant to the development of sympathetic ganglia is the demonstration that neuroendocrine cells, such as adrenal chromaffin cells and carotid body glomus cells, share a common origin with the sympathetic ganglia. Neural crest cells at the trunk level give rise to common sympathoadrenal progenitors of sympathetic neurons and chromaffin cells, while progenitors segregated from the SCG give rise to glomus cells. After separation from the sympathetic primordium, the progenitors of both chromaffin cells and glomus cells colonize the anlage of the adrenal gland and carotid body, respectively. This review highlights the biological properties of chromaffin cells and glomus cells, because, although both cell types are derivatives of sympathetic primordium, they are distinct in many respects.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan,
| |
Collapse
|
25
|
Blood pressure-lowering effect of carotid artery stenting in patients with symptomatic carotid artery stenosis. Acta Neurochir (Wien) 2014; 156:69-75. [PMID: 24241895 DOI: 10.1007/s00701-013-1928-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/21/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND In patients with symptomatic carotid artery stenosis, long-term effects of carotid artery stenting (CAS) on blood pressure (BP) changes have not been documented well. We evaluated the effects of CAS on BP and found out its predisposing factors in patients with symptomatic carotid artery stenosis. METHODS Between January 2003 and June 2012, a total of 107 patients were recruited, and all subjects met the following inclusion criteria: (1) patients underwent CAS with symptomatic carotid artery stenosis > 50%; (2) patients had clinical and radiographic data for at least 1 year of follow-up after CAS; and (3) patients had BP measurements at four different time points: pretreatment, post-treatment, 1-month follow-up, and 1-year follow-up. We evaluated the significance of the BP changes between the pretreatment BP and follow-up BPs, and determined its predisposing factors. RESULTS Compared to the mean systolic/diastolic BP value (141.0/87.4 mmHg) at the pretreatment BP, the follow-up BPs were significantly decreased after CAS (120.5/74.5, 126.2/76.9, and 129.2/79.0 mmHg at the post-treatment, the 1-month follow-up, and the 1-year follow-up, respectively [p < 0.01]). The location of the stenosis (odds ratio = 1.856, 95% confidence interval, 1.388 to 5.589; p = 0.003) and hypertension (odds ratio = 1.627, 95% confidence interval, 1.101 to 3.757; p = 0.014) were independent predisposing factors for BP-lowering effects of CAS on multivariate analysis. CONCLUSIONS For patients with symptomatic carotid artery stenosis, CAS might have a BP-lowering effect at the 1-year follow-up, especially in patients with hypertension or the stenosis at body lesions.
Collapse
|
26
|
Rana MS, Sizarov A, Christoffels VM, Moorman AFM. Development of the human aortic arch system captured in an interactive three-dimensional reference model. Am J Med Genet A 2013; 164A:1372-83. [PMID: 23613216 DOI: 10.1002/ajmg.a.35881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 12/31/2012] [Indexed: 11/12/2022]
Abstract
Variations and mutations in the human genome, such as 22q11.2 microdeletion, can increase the risk for congenital defects, including aortic arch malformations. Animal models are increasingly expanding our molecular and genetic insights into aortic arch development. However, in order to justify animal-to-human extrapolations, a human morphological, and molecular reference model would be of great value, but is currently lacking. Here, we present interactive three-dimensional reconstructions of the developing human aortic arch system, supplemented with the protein distribution of developmental markers for patterning and growth, including T-box transcription factor TBX1, a major candidate for the phenotypes found in patients with the 22q11.2 microdeletion. These reconstructions and expression data facilitate unbiased interpretations, and reveal previously unappreciated aspects of human aortic arch development. Based on our reconstructions and on reported congenital anomalies of the pulmonary trunk and tributaries, we postulate that the pulmonary arteries originate from the aortic sac, rather than from the sixth pharyngeal arch arteries. Similar to mouse, TBX1 is expressed in pharyngeal mesenchyme and epithelia. The endothelium of the pharyngeal arch arteries is largely negative for TBX1 and family member TBX2 but expresses neural crest marker AP2α, which gradually decreases with ongoing development of vascular smooth muscle. At early stages, the pharyngeal arch arteries, aortic sac, and the dorsal aortae in particular were largely negative for proliferation marker Ki67, potentially an important parameter during aortic arch system remodeling. Together, our data support current animal-to-human extrapolations and future genetic and molecular analyses using animal models of congenital heart disease. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M Sameer Rana
- Heart Failure Research Center, Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
27
|
Papangeli I, Scambler PJ. Tbx1 genetically interacts with the transforming growth factor-β/bone morphogenetic protein inhibitor Smad7 during great vessel remodeling. Circ Res 2012; 112:90-102. [PMID: 23011393 DOI: 10.1161/circresaha.112.270223] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE Growth and remodeling of the pharyngeal arch arteries are vital for the development of a mature great vessel system. Dysmorphogenesis of the fourth arch arteries can result in interruption of the aortic arch type B, typically found in DiGeorge syndrome. Tbx1 haploinsufficient embryos, which model DiGeorge syndrome, display fourth arch artery defects during formation of the vessels. Recovery from such defects is a documented yet unexplained phenotype in Tbx1 haploinsufficiency. OBJECTIVE To understand the nature of fourth arch artery growth recovery in Tbx1 haploinsufficiency and its underlying genetic control. METHODS AND RESULTS We categorized vessel phenotypes of Tbx1 heterozygotes as hypoplastic or aplastic at the conclusion of pharyngeal artery formation and compared these against the frequency of vessel defects scored at the end of great vessel development. The frequency of hypoplastic vessels decreased during embryogenesis, whereas no reduction of vessel aplasia was seen, implying recovery is attributable to remodeling of hypoplastic vessels. We showed that Smad7, an inhibitory Smad within the transforming growth factor-β pathway, is regulated by Tbx1, is required for arch artery remodeling, and genetically interacts with Tbx1 in this process. Tbx1 and Tbx1;Smad7 haploinsufficiency affected several remodeling processes; however, concurrent haploinsufficiency particularly impacted on the earliest stage of vascular smooth muscle cell vessel coverage and subsequent fibronectin deposition. Conditional reconstitution of Smad7 with a Tbx1Cre driver indicated that the interaction between the 2 genes is cell autonomous. CONCLUSIONS Tbx1 acts upstream of Smad7 controlling vascular smooth muscle and extracellular matrix investment of the fourth arch artery.
Collapse
|
28
|
Geyer SH, Weninger WJ. Some mice feature 5th pharyngeal arch arteries and double-lumen aortic arch malformations. Cells Tissues Organs 2012; 196:90-8. [PMID: 22287557 DOI: 10.1159/000330789] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2011] [Indexed: 01/21/2023] Open
Abstract
A 5th pair of pharyngeal arch arteries (PAAs) has never been identified with certainty in mice. Murines in general are considered to not develop a 5th pair. If true, the significance of the mouse as a model for researching the genesis of malformations of the great intrathoracic arteries is limited. We aimed to investigate whether mouse embryos develop a 5th pair of PAAs and to identify malformations known to be caused by defective remodelling of the 5th PAAs. We employed the high-resolution episcopic microscopy method for creating digital volume data and three-dimensional (3D) computer models of the great intrathoracic arteries of 30 mouse embryos from days 12-12.5 post conception and 180 mouse fetuses from days 14.5 and 15.5 post conception. The 3D models of the fetuses were screened for the presence of a double-lumen aortic arch malformation. We identified such a malformation in 1 fetus. The 3D models of the embryos were analysed for the presence of 5th PAAs. Six of the 30 embryos (20%) showed a 5th PAA bilaterally, and an additional 9 (30%) showed a 5th PAA unilaterally. Our results prove that some mice do develop a 5th pair of PAAs. They also show that malformations which occur rarely in humans and result from defective remodelling of the left 5th PAA can be identified in mice as well. Thus, the mouse does represent an excellent model for researching the mechanisms driving PAA remodelling and the genesis of malformations of the great intrathoracic arteries.
Collapse
Affiliation(s)
- Stefan H Geyer
- Integrative Morphology Group, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
29
|
Suh DC, Kim JL, Kim EH, Kim JK, Shin JH, Hyun DH, Lee HY, Lee DH, Kim JS. Carotid baroreceptor reaction after stenting in 2 locations of carotid bulb lesions of different embryologic origin. AJNR Am J Neuroradiol 2012; 33:977-81. [PMID: 22268083 DOI: 10.3174/ajnr.a2891] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The carotid bulb is innervated by the sinus nerve of Hering, a branch of the glossopharyngeal nerve, derived from the third pharyngeal arch. The aim of this study was to determine the frequency, predictors, and outcome of the carotid BR after carotid stent placement according to the location of the plaque lesion. MATERIALS AND METHODS Atherosclerotic carotid plaques of apical versus body lesions were prospectively analyzed in 95 consecutive patients who underwent carotid stent placement. Patients with hypertension after stent placement were excluded, and transient (<3 hours) and prolonged (3-24 hours) BR, together with AEs such as strokes and death, were assessed in the 2 lesion locations (apical versus body). Other factors known to affect the carotid baroreceptor were also investigated, and the results were analyzed by χ(2) or Mann-Whitney U tests. RESULTS Transient BR occurred in 30% of apical lesions in contrast to 70% of body lesions (P = .001). Transient BR showed a significant relationship to lesion location (P = .001), occurring most frequently in body lesions, and to the distance of maximum stenosis from the ICA ostium (P = .001). Hyperperfusion and AE rates (P = .076) in 1 month occurred more frequently in apical lesions. CONCLUSIONS The frequency of transient BR after carotid stent placement was lower in the apical region of the carotid bulb. Different cardiovascular disturbances after carotid stent placement can be attributed to anatomically different areas of the carotid bulb.
Collapse
Affiliation(s)
- D C Suh
- Department of Radiology, Asan Medical Center, University of Ulsan, College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul 138-736, Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Diman NYSG, Remacle S, Bertrand N, Picard JJ, Zaffran S, Rezsohazy R. A retinoic acid responsive Hoxa3 transgene expressed in embryonic pharyngeal endoderm, cardiac neural crest and a subdomain of the second heart field. PLoS One 2011; 6:e27624. [PMID: 22110697 PMCID: PMC3217993 DOI: 10.1371/journal.pone.0027624] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 10/20/2011] [Indexed: 11/19/2022] Open
Abstract
A transgenic mouse line harbouring a β-galacdosidase reporter gene controlled by the proximal 2 kb promoter of Hoxa3 was previously generated to investigate the regulatory cues governing Hoxa3 expression in the mouse. Examination of transgenic embryos from embryonic day (E) 8.0 to E15.5 revealed regionally restricted reporter activity in the developing heart. Indeed, transgene expression specifically delineated cells from three distinct lineages: a subpopulation of the second heart field contributing to outflow tract myocardium, the cardiac neural crest cells and the pharyngeal endoderm. Manipulation of the Retinoic Acid (RA) signaling pathway showed that RA is required for correct expression of the transgene. Therefore, this transgenic line may serve as a cardiosensor line of particular interest for further analysis of outflow tract development.
Collapse
Affiliation(s)
- Nata Y. S.-G. Diman
- Molecular and Cellular Animal Embryology group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Sophie Remacle
- Molecular and Cellular Animal Embryology group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Nicolas Bertrand
- UMR910, Aix-Marseille University, Marseille, France
- Medical Genetics and Functional Genomics, Inserm UMR_S910, Marseille, France
| | - Jacques J. Picard
- Faculty of Medicine, Université catholique de Louvain, Brussels, Belgium
| | - Stéphane Zaffran
- UMR910, Aix-Marseille University, Marseille, France
- Medical Genetics and Functional Genomics, Inserm UMR_S910, Marseille, France
- * E-mail: (SZ); (RR)
| | - René Rezsohazy
- Molecular and Cellular Animal Embryology group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- * E-mail: (SZ); (RR)
| |
Collapse
|
31
|
Sheehy NT, Cordes KR, White MP, Ivey KN, Srivastava D. The neural crest-enriched microRNA miR-452 regulates epithelial-mesenchymal signaling in the first pharyngeal arch. Development 2010; 137:4307-16. [PMID: 21098571 DOI: 10.1242/dev.052647] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neural crest cells (NCCs) are a subset of multipotent, migratory stem cells that populate a large number of tissues during development and are important for craniofacial and cardiac morphogenesis. Although microRNAs (miRNAs) have emerged as important regulators of development and disease, little is known about their role in NCC development. Here, we show that loss of miRNA biogenesis by NCC-specific disruption of murine Dicer results in embryos lacking craniofacial cartilaginous structures, cardiac outflow tract septation and thymic and dorsal root ganglia development. Dicer mutant embryos had reduced expression of Dlx2, a transcriptional regulator of pharyngeal arch development, in the first pharyngeal arch (PA1). miR-452 was enriched in NCCs, was sufficient to rescue Dlx2 expression in Dicer mutant pharyngeal arches, and regulated non-cell-autonomous signaling involving Wnt5a, Shh and Fgf8 that converged on Dlx2 regulation in PA1. Correspondingly, knockdown of miR-452 in vivo decreased Dlx2 expression in the mandibular component of PA1, leading to craniofacial defects. These results suggest that post-transcriptional regulation by miRNAs is required for differentiation of NCC-derived tissues and that miR-452 is involved in epithelial-mesenchymal signaling in the pharyngeal arch.
Collapse
Affiliation(s)
- Neil T Sheehy
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | | | | | | | | |
Collapse
|
32
|
Park ST, Kim JK, Yoon KH, Park SO, Park SW, Kim JS, Kim SJ, Suh DC. Atherosclerotic carotid stenoses of apical versus body lesions in high-risk carotid stenting patients. AJNR Am J Neuroradiol 2010; 31:1106-12. [PMID: 20093309 DOI: 10.3174/ajnr.a2000] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Different lesion locations in the atherosclerotic carotid bulb stenosis have not been clearly defined. We sought to evaluate 2 locations of carotid bulb stenosis in high-risk patients and to determine the relationship of each location to atherosclerotic risk factors and clinical features. MATERIALS AND METHODS Atherosclerotic carotid plaques of apical versus body lesions, defined according to the area and extent of plaque involvement, were retrospectively analyzed in 200 consecutive high-risk patients who underwent carotid stent placement because of > or =50% symptomatic stenosis. We evaluated interobserver concordance and assessed each type of lesion relative to 13 atherosclerotic risk factors, mode of symptom presentation, infarct pattern, procedure-related factors, and clinical outcomes, by univariate and multivariable logistic regression analysis. RESULTS Interobserver concordance showed good agreement for differentiating apical and body lesions (kappa = 0.745). Univariate analysis revealed that apical lesions (n = 108, 54%) were associated with pseudo-occlusion (P = .027), older age (P = .073), and alcohol intake (P = .080), whereas body lesions (n = 92, 46%) were associated with hyperlipidemia (P = .001), a wedge-shaped cortical infarct pattern (P = .057), and hyperperfusion syndrome (P = .083). Multivariable logistic regression analysis adjusted by age revealed that hyperlipidemia (P = .002; OR, 3.462; 95% CI, 1.595-7.515) and hyperperfusion (P = .026; OR, 6.727; 95% CI, 1.261-35.894) were independent predictors of body-type lesions. CONCLUSIONS Atherosclerotic carotid bulb stenosis was found to have 2 distinct locations, body and apical. Hyperlipidemia and cortical wedge-shaped infarcts were more frequently associated with body than with apical stenosis at the time of presentation.
Collapse
Affiliation(s)
- S-T Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Songpa-Gu, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Randall V, McCue K, Roberts C, Kyriakopoulou V, Beddow S, Barrett AN, Vitelli F, Prescott K, Shaw-Smith C, Devriendt K, Bosman E, Steffes G, Steel KP, Simrick S, Basson MA, Illingworth E, Scambler PJ. Great vessel development requires biallelic expression of Chd7 and Tbx1 in pharyngeal ectoderm in mice. J Clin Invest 2009; 119:3301-10. [PMID: 19855134 DOI: 10.1172/jci37561] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 08/19/2009] [Indexed: 11/17/2022] Open
Abstract
Aortic arch artery patterning defects account for approximately 20% of congenital cardiovascular malformations and are observed frequently in velocardiofacial syndrome (VCFS). In the current study, we screened for chromosome rearrangements in patients suspected of VCFS, but who lacked a 22q11 deletion or TBX1 mutation. One individual displayed hemizygous CHD7, which encodes a chromodomain protein. CHD7 haploinsufficiency is the major cause of coloboma, heart defect, atresia choanae, retarded growth and development, genital hypoplasia, and ear anomalies/deafness (CHARGE) syndrome, but this patient lacked the major diagnostic features of coloboma and choanal atresia. Because a subset of CHARGE cases also display 22q11 deletions, we explored the embryological relationship between CHARGE and VCSF using mouse models. The hallmark of Tbx1 haploinsufficiency is hypo/aplasia of the fourth pharyngeal arch artery (PAA) at E10.5. Identical malformations were observed in Chd7 heterozygotes, with resulting aortic arch interruption at later stages. Other than Tbx1, Chd7 is the only gene reported to affect fourth PAA development by haploinsufficiency. Moreover, Tbx1+/-;Chd7+/- double heterozygotes demonstrated a synergistic interaction during fourth PAA, thymus, and ear morphogenesis. We could not rescue PAA morphogenesis by restoring neural crest Chd7 expression. Rather, biallelic expression of Chd7 and Tbx1 in the pharyngeal ectoderm was required for normal PAA development.
Collapse
Affiliation(s)
- Victoria Randall
- Molecular Medicine Unit, Institute of Child Health, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|