1
|
Ghosh S, Tanbir SE, Mitra T, Roy SS. Unveiling stem-like traits and chemoresistance mechanisms in ovarian cancer cells through the TGFβ1-PITX2A/B signaling axis. Biochem Cell Biol 2024; 102:394-409. [PMID: 38976906 DOI: 10.1139/bcb-2024-0010] [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] [Indexed: 07/10/2024] Open
Abstract
Ovarian cancer (OC) is the deadliest gynecological malignancy, having a high mortality rate due to its asymptomatic nature, chemoresistance, and recurrence. However, the proper mechanistic knowledge behind these phenomena is still inadequate. Cancer recurrence is commonly observed due to cancer stem cells which also show chemoresistance. We aimed to decipher the molecular mechanism behind chemoresistance and stemness in OC. Earlier studies suggested that PITX2, a homeobox transcription factor and, its different isoforms are associated with OC progression upon regulating different signaling pathways. Moreover, they regulate the expression of drug efflux transporters in kidney and colon cancer, rendering chemoresistance properties in the tumor cell. Considering these backgrounds, we decided to look for the role of PITX2 isoforms in promoting stemness and chemoresistance in OC cells. In this study, PITX2A/B has been shown to promote stemness and to enhance the transcription of ABCB1. PITX2 has been discovered to augment ABCB1 gene expression by directly binding to its promoter. To further investigate the regulatory mechanism of PITX2 gene expression, we found that TGFβ signaling could augment the PITX2A/B expression through both SMAD and non-SMAD signaling pathways. Collectively, we conclude that TGFβ1-activated PITX2A/B induces stem-like features and chemoresistance properties in the OC cells.
Collapse
Affiliation(s)
- Sampurna Ghosh
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sk Eashayan Tanbir
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Tulika Mitra
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sib Sankar Roy
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Nilsson KH, Henning P, Wu J, Sjögren K, Lerner UH, Ohlsson C, Movérare-Skrtic S. GREM2 inactivation increases trabecular bone mass in mice. Sci Rep 2024; 14:12967. [PMID: 38839844 PMCID: PMC11153596 DOI: 10.1038/s41598-024-63439-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Osteoporosis is a common skeletal disease affecting millions of individuals world-wide, with an increased risk of fracture, and a decreased quality of life. Despite its well-known consequences, the etiology of osteoporosis and optimal treatment methods are not fully understood. Human genetic studies have identified genetic variants within the FMN2/GREM2 locus to be associated with trabecular volumetric bone mineral density (vBMD) and vertebral and forearm fractures, but not with cortical bone parameters. GREM2 is a bone morphogenetic protein (BMP) antagonist. In this study, we employed Grem2-deficient mice to investigate whether GREM2 serves as the plausible causal gene for the fracture signal at the FMN2/GREM2 locus. We observed that Grem2 is moderately expressed in bone tissue and particularly in osteoblasts. Complete Grem2 gene deletion impacted mouse survival and body growth. Partial Grem2 inactivation in Grem2+/- female mice led to increased trabecular BMD of femur and increased trabecular bone mass in tibia due to increased trabecular thickness, with an unchanged cortical thickness, as compared with wildtype littermates. Furthermore, Grem2 inactivation stimulated osteoblast differentiation, as evidenced by higher alkaline phosphatase (Alp), osteocalcin (Bglap), and osterix (Sp7) mRNA expression after BMP-2 stimulation in calvarial osteoblasts and osteoblasts from the long bones of Grem2-/- mice compared to wildtype littermates. These findings suggest that GREM2 is a possible target for novel osteoporotic treatments, to increase trabecular bone mass and prevent osteoporotic fractures.
Collapse
Affiliation(s)
- Karin H Nilsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jianyao Wu
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Klara Sjögren
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf H Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
3
|
Grau-Bové X, Subirana L, Meister L, Soubigou A, Neto A, Elek A, Naranjo S, Fornas O, Gomez-Skarmeta JL, Tena JJ, Irimia M, Bertrand S, Sebé-Pedrós A, Escriva H. An amphioxus neurula stage cell atlas supports a complex scenario for the emergence of vertebrate head mesoderm. Nat Commun 2024; 15:4550. [PMID: 38811547 PMCID: PMC11136973 DOI: 10.1038/s41467-024-48774-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
The emergence of new structures can often be linked to the evolution of novel cell types that follows the rewiring of developmental gene regulatory subnetworks. Vertebrates are characterized by a complex body plan compared to the other chordate clades and the question remains of whether and how the emergence of vertebrate morphological innovations can be related to the appearance of new embryonic cell populations. We previously proposed, by studying mesoderm development in the cephalochordate amphioxus, a scenario for the evolution of the vertebrate head mesoderm. To further test this scenario at the cell population level, we used scRNA-seq to construct a cell atlas of the amphioxus neurula, stage at which the main mesodermal compartments are specified. Our data allowed us to validate the presence of a prechordal-plate like territory in amphioxus. Additionally, the transcriptomic profile of somite cell populations supports the homology between specific territories of amphioxus somites and vertebrate cranial/pharyngeal and lateral plate mesoderm. Finally, our work provides evidence that the appearance of the specific mesodermal structures of the vertebrate head was associated to both segregation of pre-existing cell populations, and co-option of new genes for the control of myogenesis.
Collapse
Affiliation(s)
- Xavier Grau-Bové
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Lucie Subirana
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, F-66650, Banyuls-sur-Mer, France
| | - Lydvina Meister
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, F-66650, Banyuls-sur-Mer, France
| | - Anaël Soubigou
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, F-66650, Banyuls-sur-Mer, France
| | - Ana Neto
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
| | - Anamaria Elek
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Silvia Naranjo
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
| | - Oscar Fornas
- Flow Cytometry Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jose Luis Gomez-Skarmeta
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Universidad Pablo de Olavide-Junta de Andalucía, Sevilla, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Stéphanie Bertrand
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, F-66650, Banyuls-sur-Mer, France.
- Institut universitaire de France (IUF), Paris, France.
| | - Arnau Sebé-Pedrós
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| | - Hector Escriva
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, F-66650, Banyuls-sur-Mer, France.
| |
Collapse
|
4
|
Grass A, Kasajima A, Foersch S, Kriegsmann M, Brobeil A, Schmitt M, Wagner D, Poppinga J, Wiese D, Maurer E, Kirschbaum A, Muley T, Winter H, Rinke A, Gress TM, Kremer M, Evert M, Märkl B, Quaas A, Eckstein M, Tschurtschenthaler M, Klöppel G, Denkert C, Bartsch DK, Jesinghaus M. PITX2 as a Sensitive and Specific Marker of Midgut Neuroendocrine Tumors: Results from a Cohort of 1157 Primary Neuroendocrine Neoplasms. Mod Pathol 2024; 37:100442. [PMID: 38309431 DOI: 10.1016/j.modpat.2024.100442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
As neuroendocrine tumors (NETs) often present as metastatic lesions, immunohistochemical assignment to a site of origin is one of the most important tasks in their pathologic assessment. Because a fraction of NETs eludes the typical expression profiles of their primary localization, additional sensitive and specific markers are required to improve diagnostic certainty. We investigated the expression of the transcription factor Pituitary Homeobox 2 (PITX2) in a large-scale cohort of 909 NET and 248 neuroendocrine carcinomas (NEC) according to the immunoreactive score (IRS) and correlated PITX2 expression groups with general tumor groups and primary localization. PITX2 expression (all expression groups) was highly sensitive (98.1%) for midgut-derived NET, but not perfectly specific, as non-midgut NET (especially pulmonary/duodenal) were quite frequently weak or moderately positive. The specificity rose to 99.5% for a midgut origin of NET if only a strong PITX2 expression was considered, which was found in only 0.5% (one pancreatic/one pulmonary) of non-midgut NET. In metastases of midgut-derived NET, PITX2 was expressed in all cases (87.5% strong, 12.5% moderate), whereas CDX2 was negative or only weakly expressed in 31.3% of the metastases. In NEC, a fraction of cases (14%) showed a weak or moderate PITX2 expression, which was not associated with a specific tumor localization. Our study independently validates PITX2 as a very sensitive and specific immunohistochemical marker of midgut-derived NET in a very large collective of neuroendocrine neoplasms. Therefore, our data argue toward implementation into diagnostic panels applied for NET as a firstline midgut marker.
Collapse
Affiliation(s)
- Albert Grass
- Department of Pathology, Phillips University Marburg und University Hospital Marburg, Marburg, Germany
| | - Atsuko Kasajima
- Department of Pathology, Technical University of Munich, Munich, Germany
| | | | - Mark Kriegsmann
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Brobeil
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maxime Schmitt
- Department of Pathology, Phillips University Marburg und University Hospital Marburg, Marburg, Germany
| | - Daniel Wagner
- Department of Pathology, University Hospital Mainz, Mainz, Germany
| | - Jelte Poppinga
- Department of Surgery, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Dominik Wiese
- Department of Surgery, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Elisabeth Maurer
- Department of Surgery, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Andreas Kirschbaum
- Department of Surgery, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany; Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany; Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany; Department of Thoracic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Anja Rinke
- Department of Gastroenterology, Endocrinology and Infectious Diseases, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology and Infectious Diseases, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Markus Kremer
- Institute of Pathology, Städtisches Klinikum München, Munich, Germany
| | - Matthias Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - Bruno Märkl
- Institute of Pathology, University Hospital Augsburg, Augsburg, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Markus Eckstein
- Department of Pathology, University Hospital Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus Tschurtschenthaler
- Institute for Translational Cancer Research, German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Günter Klöppel
- Department of Pathology, Technical University of Munich, Munich, Germany
| | - Carsten Denkert
- Department of Pathology, Phillips University Marburg und University Hospital Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of Surgery, Phillips University Marburg and University Hospital Marburg, Marburg, Germany
| | - Moritz Jesinghaus
- Department of Pathology, Phillips University Marburg und University Hospital Marburg, Marburg, Germany.
| |
Collapse
|
5
|
Gabriel GC, Wu YL, Lo CW. Establishment of Cardiac Laterality. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:167-183. [PMID: 38884711 DOI: 10.1007/978-3-031-44087-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Formation of the vertebrate heart with its complex arterial and venous connections is critically dependent on patterning of the left-right axis during early embryonic development. Abnormalities in left-right patterning can lead to a variety of complex life-threatening congenital heart defects. A highly conserved pathway responsible for left-right axis specification has been uncovered. This pathway involves initial asymmetric activation of a nodal signaling cascade at the embryonic node, followed by its propagation to the left lateral plate mesoderm and activation of left-sided expression of the Pitx2 transcription factor specifying visceral organ asymmetry. Intriguingly, recent work suggests that cardiac laterality is encoded by intrinsic cell and tissue chirality independent of Nodal signaling. Thus, Nodal signaling may be superimposed on this intrinsic chirality, providing additional instructive cues to pattern cardiac situs. The impact of intrinsic chirality and the perturbation of left-right patterning on myofiber organization and cardiac function warrants further investigation. We summarize recent insights gained from studies in animal models and also some human clinical studies in a brief overview of the complex processes regulating cardiac asymmetry and their impact on cardiac function and the pathogenesis of congenital heart defects.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yijen L Wu
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Chacon-Camacho OF, Arce-Gonzalez R, Sanchez-de la Rosa F, Urióstegui-Rojas A, Hofmann-Blancas ME, Mata-Flores F, Zenteno JC. Genetic Aspects of Glaucoma: An Updated Review. Curr Mol Med 2024; 24:1231-1249. [PMID: 37272463 DOI: 10.2174/1566524023666230602143617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 06/06/2023]
Abstract
Glaucoma is a group of diverse diseases characterized by cupping of the optic nerve head due to the loss of retinal ganglion cells. It is the most common cause of irreversible blindness throughout the world; therefore, its timely diagnosis and early detection through an ophthalmological examination are very important. We, herein, present the information on the epidemiology, pathophysiology, clinical diagnosis, and treatment of glaucoma. We also emphasize the investigations of the last decades that have allowed identifying numerous genes and susceptibility genetic factors. We have also described in detail the genes whose mutations cause or contribute to the development of the disease.
Collapse
Affiliation(s)
- Oscar Francisco Chacon-Camacho
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
- Laboratorio 5 Edificio A-4, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rocio Arce-Gonzalez
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Andrés Urióstegui-Rojas
- Department of Integral Ophthalmology, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Felipe Mata-Flores
- Department of Glaucoma, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | - Juan Carlos Zenteno
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
- Biochemistry Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
7
|
Attico E, Galaverni G, Torello A, Bianchi E, Bonacorsi S, Losi L, Manfredini R, Lambiase A, Rama P, Pellegrini G. Comparison between Cultivated Oral Mucosa and Ocular Surface Epithelia for COMET Patients Follow-Up. Int J Mol Sci 2023; 24:11522. [PMID: 37511281 PMCID: PMC10380900 DOI: 10.3390/ijms241411522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Total bilateral Limbal Stem Cell Deficiency is a pathologic condition of the ocular surface due to the loss of corneal stem cells. Cultivated oral mucosa epithelial transplantation (COMET) is the only autologous successful treatment for this pathology in clinical application, although abnormal peripheric corneal vascularization often occurs. Properly characterizing the regenerated ocular surface is needed for a reliable follow-up. So far, the univocal identification of transplanted oral mucosa has been challenging. Previously proposed markers were shown to be co-expressed by different ocular surface epithelia in a homeostatic or perturbated environment. In this study, we compared the transcriptome profile of human oral mucosa, limbal and conjunctival cultured holoclones, identifying Paired Like Homeodomain 2 (PITX2) as a new marker that univocally distinguishes the transplanted oral tissue from the other epithelia. We validated PITX2 at RNA and protein levels to investigate 10-year follow-up corneal samples derived from a COMET-treated aniridic patient. Moreover, we found novel angiogenesis-related factors that were differentially expressed in the three epithelia and instrumental in explaining the neovascularization in COMET-treated patients. These results will support the follow-up analysis of patients transplanted with oral mucosa and provide new tools to understand the regeneration mechanism of transplanted corneas.
Collapse
Affiliation(s)
- Eustachio Attico
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Galaverni
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Andrea Torello
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
- Holostem Terapie Avanzate s.r.l., 41125 Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Susanna Bonacorsi
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Lorena Losi
- Unit of Pathology, Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | - Paolo Rama
- SC Ophathalmology, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy
| | - Graziella Pellegrini
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, 41125 Modena, Italy
- Holostem Terapie Avanzate s.r.l., 41125 Modena, Italy
| |
Collapse
|
8
|
Forrest K, Barricella AC, Pohar SA, Hinman AM, Amack JD. Understanding laterality disorders and the left-right organizer: Insights from zebrafish. Front Cell Dev Biol 2022; 10:1035513. [PMID: 36619867 PMCID: PMC9816872 DOI: 10.3389/fcell.2022.1035513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Vital internal organs display a left-right (LR) asymmetric arrangement that is established during embryonic development. Disruption of this LR asymmetry-or laterality-can result in congenital organ malformations. Situs inversus totalis (SIT) is a complete concordant reversal of internal organs that results in a low occurrence of clinical consequences. Situs ambiguous, which gives rise to Heterotaxy syndrome (HTX), is characterized by discordant development and arrangement of organs that is associated with a wide range of birth defects. The leading cause of health problems in HTX patients is a congenital heart malformation. Mutations identified in patients with laterality disorders implicate motile cilia in establishing LR asymmetry. However, the cellular and molecular mechanisms underlying SIT and HTX are not fully understood. In several vertebrates, including mouse, frog and zebrafish, motile cilia located in a "left-right organizer" (LRO) trigger conserved signaling pathways that guide asymmetric organ development. Perturbation of LRO formation and/or function in animal models recapitulates organ malformations observed in SIT and HTX patients. This provides an opportunity to use these models to investigate the embryological origins of laterality disorders. The zebrafish embryo has emerged as an important model for investigating the earliest steps of LRO development. Here, we discuss clinical characteristics of human laterality disorders, and highlight experimental results from zebrafish that provide insights into LRO biology and advance our understanding of human laterality disorders.
Collapse
Affiliation(s)
- Kadeen Forrest
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Alexandria C. Barricella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Sonny A. Pohar
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Anna Maria Hinman
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, United States
| |
Collapse
|
9
|
Prieto JM, Wang AW, Halbach J, Cauvi DM, Day JMD, Gembicky M, Ghassemian M, Quehenberger O, Kling K, Ignacio R, DeMaio A, Bickler SW. Elemental, fatty acid, and protein composition of appendicoliths. Sci Rep 2022; 12:19764. [PMID: 36396724 PMCID: PMC9671961 DOI: 10.1038/s41598-022-21397-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Appendicoliths are commonly found obstructing the lumen of the appendix at the time of appendectomy. To identify factors that might contribute to their formation we investigated the composition of appendicoliths using laser ablation inductively coupled plasma mass spectroscopy, gas chromatography, polarized light microscopy, X-ray crystallography and protein mass spectroscopy. Forty-eight elements, 32 fatty acids and 109 human proteins were identified within the appendicoliths. The most common elements found in appendicoliths are calcium and phosphorus, 11.0 ± 6.0 and 8.2 ± 4.2% weight, respectively. Palmitic acid (29.7%) and stearate (21.3%) are the most common fatty acids. Some stearate is found in crystalline form-identifiable by polarized light microscopy and confirmable by X-ray crystallography. Appendicoliths have an increased ratio of omega-6 to omega-3 fatty acids (ratio 22:1). Analysis of 16 proteins common to the appendicoliths analyzed showed antioxidant activity and neutrophil functions (e.g. activation and degranulation) to be the most highly enriched pathways. Considered together, these preliminary findings suggest oxidative stress may have a role in appendicolith formation. Further research is needed to determine how dietary factors such as omega-6 fatty acids and food additives, redox-active metals and the intestinal microbiome interact with genetic factors to predispose to appendicolith formation.
Collapse
Affiliation(s)
- James M Prieto
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA
- Naval Medical Center San Diego, San Diego, CA, USA
| | - Andrew W Wang
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA
- Naval Medical Center San Diego, San Diego, CA, USA
| | - Jonathan Halbach
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA
- Naval Medical Center San Diego, San Diego, CA, USA
| | - David M Cauvi
- Naval Medical Center San Diego, San Diego, CA, USA
- Center for Investigations of Health and Education Disparities, University of California San Diego, La Jolla, CA, USA
| | - James M D Day
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Milan Gembicky
- Crystallography Facility, Department of Chemistry & Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA, USA
| | | | - Karen Kling
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA
- Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Drive #0739, La Jolla, CA, 92093-0739, USA
| | - Romeo Ignacio
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA
- Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Drive #0739, La Jolla, CA, 92093-0739, USA
| | - Antonio DeMaio
- Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Drive #0739, La Jolla, CA, 92093-0739, USA
- Center for Investigations of Health and Education Disparities, University of California San Diego, La Jolla, CA, USA
| | - Stephen W Bickler
- Division of Pediatric Surgery, Rady Children's Hospital, San Diego, CA, USA.
- Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Drive #0739, La Jolla, CA, 92093-0739, USA.
- Center for Investigations of Health and Education Disparities, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Agostini C, Bühler A, Antico Calderone A, Aadepu N, Herder C, Loosli F, Carl M. Conserved and diverged asymmetric gene expression in the brain of teleosts. Front Cell Dev Biol 2022; 10:1005776. [PMID: 36211473 PMCID: PMC9532764 DOI: 10.3389/fcell.2022.1005776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Morphological left-right brain asymmetries are universal phenomena in animals. These features have been studied for decades, but the functional relevance is often unclear. Studies from the zebrafish dorsal diencephalon on the genetics underlying the establishment and function of brain asymmetries have uncovered genes associated with the development of functional brain asymmetries. To gain further insights, comparative studies help to investigate the emergence of asymmetries and underlying genetics in connection to functional adaptation. Evolutionarily distant isogenic medaka inbred lines, that show divergence of complex traits such as morphology, physiology and behavior, are a valuable resource to investigate intra-species variations in a given trait of interest. For a detailed study of asymmetry in the medaka diencephalon we generated molecular probes of ten medaka genes that are expressed asymmetrically in the zebrafish habenulae and pineal complex. We find expression of eight genes in the corresponding brain areas of medaka with differences in the extent of left-right asymmetry compared to zebrafish. Our marker gene analysis of the diverged medaka inbred strains revealed marked inter-strain size differences of the respective expression domains in the parapineal and the habenulae, which we hypothesize may result from strain-specific gene loss. Thus, our analysis reveals both inter-species differences but also intra-species plasticity of gene expression in the teleost dorsal diencephalon. These findings are a starting point showing the potential to identify the genetics underlying the emergence and modulations of asymmetries. They are also the prerequisite to examine whether variance in habenular gene expression may cause variation of behavioral traits.
Collapse
Affiliation(s)
- Carolina Agostini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Anja Bühler
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | | | - Narendar Aadepu
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Karlsruhe, Germany
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Cathrin Herder
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Felix Loosli
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology, Karlsruhe, Germany
- *Correspondence: Felix Loosli, ; Matthias Carl,
| | - Matthias Carl
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- *Correspondence: Felix Loosli, ; Matthias Carl,
| |
Collapse
|
11
|
Derrick CJ, Santos-Ledo A, Eley L, Paramita IA, Henderson DJ, Chaudhry B. Sequential action of JNK genes establishes the embryonic left-right axis. Development 2022; 149:274898. [PMID: 35352808 PMCID: PMC9148569 DOI: 10.1242/dev.200136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/09/2022] [Indexed: 12/22/2022]
Abstract
The establishment of the left-right axis is crucial for the placement, morphogenesis and function of internal organs. Left-right specification is proposed to be dependent on cilia-driven fluid flow in the embryonic node. Planar cell polarity (PCP) signalling is crucial for patterning of nodal cilia, yet downstream effectors driving this process remain elusive. We have examined the role of the JNK gene family, a proposed downstream component of PCP signalling, in the development and function of the zebrafish node. We show jnk1 and jnk2 specify length of nodal cilia, generate flow in the node and restrict southpaw to the left lateral plate mesoderm. Moreover, loss of asymmetric southpaw expression does not result in disturbances to asymmetric organ placement, supporting a model in which nodal flow may be dispensable for organ laterality. Later, jnk3 is required to restrict pitx2c expression to the left side and permit correct endodermal organ placement. This work uncovers multiple roles for the JNK gene family acting at different points during left-right axis establishment. It highlights extensive redundancy and indicates JNK activity is distinct from the PCP signalling pathway.
Collapse
Affiliation(s)
- Christopher J Derrick
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Adrian Santos-Ledo
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Lorraine Eley
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Isabela Andhika Paramita
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Deborah J Henderson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
12
|
Akinaga K, Azumi Y, Mogi K, Toyoizumi R. Stage-dependent sequential organization of nascent smooth muscle cells and its implications for the gut coiling morphogenesis in Xenopus larva. ZOOLOGY 2021; 146:125905. [PMID: 33631602 DOI: 10.1016/j.zool.2021.125905] [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: 11/17/2020] [Revised: 01/25/2021] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
In vertebrates, gut coiling proceeds left-right asymmetrically during expansion of the gastrointestinal tract with highly organized muscular structures facilitating peristalsis. In this report, we explored the mechanisms of larval gut coiling morphogenesis relevant to its nascent smooth muscle cells using highly transparent Xenopus early larvae. First, to visualize the dynamics of intestinal smooth muscle cells, whole-mount specimens were immunostained with anti-smooth muscle-specific actin (SM-actin) antibody. We found that the nascent gut of Xenopus early larvae gradually expands the SM-actin-positive region in a stage-dependent manner. Transverse orientation of smooth muscle cells was first established, and next, the cellular longitudinal orientation along the gut axis was followed to make a meshwork of the contractile cells. Finally, anisotropic torsion by the smooth muscle cells was generated in the center of gut coiling, suggesting that twisting force might be involved in the late phase of coiling morphogenesis of the gut. Administration of S-(-)-Blebbistatin to attenuate the actomyosin contraction in vivo resulted in cancellation of coiling of the gut. Development of decapitation embryos, trunk 'torso' explants, and gut-only explants revealed that initial coiling of the gut proceeds without interactions with the other parts of the body including the central nervous system. We newly developed an in vitro model to assess the gut coiling morphogenesis, indicating that coiling pattern of the nascent Xenopus gut is partially gut-autonomous. Using this gut explant culture technique, inhibition of actomyosin contraction was performed by administrating either actin polymerization inhibitor, myosin light chain kinase inhibitor, or calmodulin antagonist. All of these reagents decreased the extent of gut coiling morphogenesis in vitro. Taken together, these results suggest that the contraction force generated by actomyosin-rich intestinal smooth muscle cells during larval stages is essential for the normal coiling morphogenesis of this muscular tubular organ.
Collapse
Affiliation(s)
- Kaoru Akinaga
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan
| | - Yoshitaka Azumi
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan; Research Institute for Integrated Science, Kanagawa University, Japan
| | - Kazue Mogi
- Research Institute for Integrated Science, Kanagawa University, Japan
| | - Ryuji Toyoizumi
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan; Research Institute for Integrated Science, Kanagawa University, Japan.
| |
Collapse
|
13
|
Abstract
Cardiac development is a complex developmental process that is initiated soon after gastrulation, as two sets of precardiac mesodermal precursors are symmetrically located and subsequently fused at the embryonic midline forming the cardiac straight tube. Thereafter, the cardiac straight tube invariably bends to the right, configuring the first sign of morphological left–right asymmetry and soon thereafter the atrial and ventricular chambers are formed, expanded and progressively septated. As a consequence of all these morphogenetic processes, the fetal heart acquired a four-chambered structure having distinct inlet and outlet connections and a specialized conduction system capable of directing the electrical impulse within the fully formed heart. Over the last decades, our understanding of the morphogenetic, cellular, and molecular pathways involved in cardiac development has exponentially grown. Multiples aspects of the initial discoveries during heart formation has served as guiding tools to understand the etiology of cardiac congenital anomalies and adult cardiac pathology, as well as to enlighten novels approaches to heal the damaged heart. In this review we provide an overview of the complex cellular and molecular pathways driving heart morphogenesis and how those discoveries have provided new roads into the genetic, clinical and therapeutic management of the diseased hearts.
Collapse
|
14
|
Wei L, Al Oustah A, Blader P, Roussigné M. Notch signaling restricts FGF pathway activation in parapineal cells to promote their collective migration. eLife 2019; 8:46275. [PMID: 31498774 PMCID: PMC6733574 DOI: 10.7554/elife.46275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/18/2019] [Indexed: 12/16/2022] Open
Abstract
Coordinated migration of cell collectives is important during embryonic development and relies on cells integrating multiple mechanical and chemical cues. Recently, we described that focal activation of the FGF pathway promotes the migration of the parapineal in the zebrafish epithalamus. How FGF activity is restricted to leading cells in this system is, however, unclear. Here, we address the role of Notch signaling in modulating FGF activity within the parapineal. While Notch loss-of-function results in an increased number of parapineal cells activating the FGF pathway, global activation of Notch signaling decreases it; both contexts result in defects in parapineal migration and specification. Decreasing or increasing FGF signaling in a Notch loss-of-function context respectively rescues or aggravates parapineal migration defects without affecting parapineal cells specification. We propose that Notch signaling controls the migration of the parapineal through its capacity to restrict FGF pathway activation to a few leading cells.
Collapse
Affiliation(s)
- Lu Wei
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Amir Al Oustah
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Patrick Blader
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Myriam Roussigné
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| |
Collapse
|
15
|
Wang H, Holland PWH, Takahashi T. Gene profiling of head mesoderm in early zebrafish development: insights into the evolution of cranial mesoderm. EvoDevo 2019; 10:14. [PMID: 31312422 PMCID: PMC6612195 DOI: 10.1186/s13227-019-0128-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 06/26/2019] [Indexed: 11/10/2022] Open
Abstract
Background The evolution of the head was one of the key events that marked the transition from invertebrates to vertebrates. With the emergence of structures such as eyes and jaws, vertebrates evolved an active and predatory life style and radiated into diversity of large-bodied animals. These organs are moved by cranial muscles that derive embryologically from head mesoderm. Compared with other embryonic components of the head, such as placodes and cranial neural crest cells, our understanding of cranial mesoderm is limited and is restricted to few species. Results Here, we report the expression patterns of key genes in zebrafish head mesoderm at very early developmental stages. Apart from a basic anterior–posterior axis marked by a combination of pitx2 and tbx1 expression, we find that most gene expression patterns are poorly conserved between zebrafish and chick, suggesting fewer developmental constraints imposed than in trunk mesoderm. Interestingly, the gene expression patterns clearly show the early establishment of medial–lateral compartmentalisation in zebrafish head mesoderm, comprising a wide medial zone flanked by two narrower strips. Conclusions In zebrafish head mesoderm, there is no clear molecular regionalisation along the anteroposterior axis as previously reported in chick embryos. In contrast, the medial–lateral regionalisation is formed at early developmental stages. These patterns correspond to the distinction between paraxial mesoderm and lateral plate mesoderm in the trunk, suggesting a common groundplan for patterning head and trunk mesoderm. By comparison of these expression patterns to that of amphioxus homologues, we argue for an evolutionary link between zebrafish head mesoderm and amphioxus anteriormost somites. Electronic supplementary material The online version of this article (10.1186/s13227-019-0128-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huijia Wang
- 1Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Peter W H Holland
- 2Department of Zoology, University of Oxford, Zoology Research and Administration Building, 11a Mansfield Road, Oxford, OX1 3SZ UK
| | - Tokiharu Takahashi
- 1Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT UK
| |
Collapse
|
16
|
Orlova E, Yeh A, Shi M, Firek B, Ranganathan S, Whitcomb DC, Finegold DN, Ferrell RE, Barmada MM, Marazita ML, Hinds DA, Shaffer JR, Morowitz MJ. Genetic association and differential expression of PITX2 with acute appendicitis. Hum Genet 2019; 138:37-47. [PMID: 30392061 PMCID: PMC6514078 DOI: 10.1007/s00439-018-1956-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022]
Abstract
Appendicitis affects 9% of Americans and is the most common diagnosis requiring hospitalization of both children and adults. We performed a genome-wide association study of self-reported appendectomy with 18,773 affected adults and 114,907 unaffected adults of European American ancestry. A significant association with appendectomy was observed at 4q25 near the gene PITX2 (rs2129979, p value = 8.82 × 10-14) and was replicated in an independent sample of Caucasians (59 affected, 607 unaffected; p value = 0.005). Meta-analysis of the associated variant across our two cohorts and cohorts from Iceland and the Netherlands (in which this association had previously been reported) showed strong cumulative evidence of association (OR = 1.12; 95% CI 1.09-1.14; p value = 1.81 × 10-23) and some evidence for effect heterogeneity (p value = 0.03). Eight other loci were identified at suggestive significance in the discovery GWAS. Associations were followed up by measuring gene expression across resected appendices with varying levels of inflammation (N = 75). We measured expression of 27 genes based on physical proximity to the GWAS signals, evidence of being targeted by eQTLs near the signals according to RegulomeDB (score = 1), or both. Four of the 27 genes (including PITX2) showed significant evidence (p values < 0.0033) of differential expression across categories of appendix inflammation. An additional ten genes showed nominal evidence (p value < 0.05) of differential expression, which, together with the significant genes, is more than expected by chance (p value = 6.6 × 10-12). PITX2 impacts morphological development of intestinal tissue, promotes an anti-oxidant response, and its expression correlates with levels of intestinal bacteria and colonic inflammation. Further studies of the role of PITX2 in appendicitis are warranted.
Collapse
Affiliation(s)
- Ekaterina Orlova
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
- Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Andrew Yeh
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Min Shi
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Sarangarajan Ranganathan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - David C Whitcomb
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
- Department of Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Department of Cell Biology and Molecular Physiology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - David N Finegold
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
| | - Robert E Ferrell
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
| | - M Michael Barmada
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
| | - Mary L Marazita
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA
- Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - John R Shaffer
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, 130 De Soto Street, 3131 Parran Hall, Pittsburgh, PA, 15261, USA.
- Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Michael J Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- Faculty Pavilion 7th Floor, Children's Hospital of Pittsburgh of UPMC, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
17
|
Li X, Zhang C, Gong T, Ni X, Li J, Zhan D, Liu M, Song L, Ding C, Xu J, Zhen B, Wang Y, Qin J. A time-resolved multi-omic atlas of the developing mouse stomach. Nat Commun 2018; 9:4910. [PMID: 30464175 PMCID: PMC6249217 DOI: 10.1038/s41467-018-07463-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023] Open
Abstract
The mammalian stomach is structurally highly diverse and its organ functionality critically depends on a normal embryonic development. Although there have been several studies on the morphological changes during stomach development, a system-wide analysis of the underlying molecular changes is lacking. Here, we present a comprehensive, temporal proteome and transcriptome atlas of the mouse stomach at multiple developmental stages. Quantitative analysis of 12,108 gene products allows identifying three distinct phases based on changes in proteins and RNAs and the gain of stomach functions on a longitudinal time scale. The transcriptome indicates functionally important isoforms relevant to development and identifies several functionally unannotated novel splicing junction transcripts that we validate at the peptide level. Importantly, many proteins differentially expressed in stomach development are also significantly overexpressed in diffuse-type gastric cancer. Overall, our study provides a resource to understand stomach development and its connection to gastric cancer tumorigenesis.
Collapse
Affiliation(s)
- Xianju Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Chunchao Zhang
- Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tongqing Gong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Xiaotian Ni
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China.,Department of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jin'e Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China.,Department of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China
| | - Jianming Xu
- Department of Gastrointestinal Oncology, Affiliated Hospital Cancer Center, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Bei Zhen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China. .,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China. .,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Collins MM, Maischein HM, Dufourcq P, Charpentier M, Blader P, Stainier DY. Pitx2c orchestrates embryonic axis extension via mesendodermal cell migration. eLife 2018; 7:34880. [PMID: 29952749 PMCID: PMC6023614 DOI: 10.7554/elife.34880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
Pitx2c, a homeodomain transcription factor, is classically known for its left-right patterning role. However, an early wave of pitx2 expression occurs at the onset of gastrulation in several species, indicating a possible earlier role that remains relatively unexplored. Here we show that in zebrafish, maternal-zygotic (MZ) pitx2c mutants exhibit a shortened body axis indicative of convergence and extension (CE) defects. Live imaging reveals that MZpitx2c mutants display less persistent mesendodermal migration during late stages of gastrulation. Transplant data indicate that Pitx2c functions cell non-autonomously to regulate this cell behavior by modulating cell shape and protrusive activity. Using transcriptomic analyses and candidate gene approaches, we identify transcriptional changes in components of the chemokine-ECM-integrin dependent mesendodermal migration network. Together, our results define pathways downstream of Pitx2c that are required during early embryogenesis and reveal novel functions for Pitx2c as a regulator of morphogenesis.
Collapse
Affiliation(s)
- Michelle M Collins
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Pascale Dufourcq
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université Toulouse III - Paul Sabatier, CNRS, Toulouse, France
| | | | - Patrick Blader
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université Toulouse III - Paul Sabatier, CNRS, Toulouse, France
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
19
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
20
|
Seifi M, Walter MA. Axenfeld-Rieger syndrome. Clin Genet 2018; 93:1123-1130. [PMID: 28972279 DOI: 10.1111/cge.13148] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/23/2017] [Accepted: 09/26/2017] [Indexed: 12/29/2022]
Abstract
Axenfeld-Rieger syndrome (ARS) is a clinically and genetically heterogeneous group of developmental disorders affecting primarily the anterior segment of the eye, often leading to secondary glaucoma. Patients with ARS may also present with systemic changes, including dental defects, mild craniofacial dysmorphism, and umbilical anomalies. ARS is inherited in an autosomal-dominant fashion; the underlying defect in 40% of patients is mutations in PITX2 or FOXC1. Here, an overview of the clinical spectrum of ARS is provided. As well, the known underlying genetic defects, clinical diagnostic possibilities, genetic counseling and treatments of ARS are discussed in detail.
Collapse
Affiliation(s)
- M Seifi
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Alberta, Edmonton, Canada
| | - M A Walter
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Alberta, Edmonton, Canada
| |
Collapse
|
21
|
Multiple Roles of Pitx2 in Cardiac Development and Disease. J Cardiovasc Dev Dis 2017; 4:jcdd4040016. [PMID: 29367545 PMCID: PMC5753117 DOI: 10.3390/jcdd4040016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Cardiac development is a complex morphogenetic process initiated as bilateral cardiogenic mesoderm is specified at both sides of the gastrulating embryo. Soon thereafter, these cardiogenic cells fuse at the embryonic midline configuring a symmetrical linear cardiac tube. Left/right bilateral asymmetry is first detected in the forming heart as the cardiac tube bends to the right, and subsequently, atrial and ventricular chambers develop. Molecular signals emanating from the node confer distinct left/right signalling pathways that ultimately lead to activation of the homeobox transcription factor Pitx2 in the left side of distinct embryonic organ anlagen, including the developing heart. Asymmetric expression of Pitx2 has therefore been reported during different cardiac developmental stages, and genetic deletion of Pitx2 provided evidence of key regulatory roles of this transcription factor during cardiogenesis and thus congenital heart diseases. More recently, impaired Pitx2 function has also been linked to arrhythmogenic processes, providing novel roles in the adult heart. In this manuscript, we provide a state-of-the-art review of the fundamental roles of Pitx2 during cardiogenesis, arrhythmogenesis and its contribution to congenital heart diseases.
Collapse
|
22
|
Functional characterization of zebrafish orthologs of the human Beta 3-Glucosyltransferase B3GLCT gene mutated in Peters Plus Syndrome. PLoS One 2017; 12:e0184903. [PMID: 28926587 PMCID: PMC5604996 DOI: 10.1371/journal.pone.0184903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/01/2017] [Indexed: 12/25/2022] Open
Abstract
Peters Plus Syndrome (PPS) is a rare autosomal recessive disease characterized by ocular defects, short stature, brachydactyly, characteristic facial features, developmental delay and other highly variable systemic defects. Classic PPS is caused by loss-of-function mutations in the B3GLCT gene encoding for a β3-glucosyltransferase that catalyzes the attachment of glucose via a β1–3 glycosidic linkage to O-linked fucose on thrombospondin type 1 repeats (TSRs). B3GLCT was shown to participate in a non-canonical ER quality control mechanism; however, the exact molecular processes affected in PPS are not well understood. Here we report the identification and characterization of two zebrafish orthologs of the human B3GLCT gene, b3glcta and b3glctb. The b3glcta and b3glctb genes encode for 496-aa and 493-aa proteins with 65% and 57% identity to human B3GLCT, respectively. Expression studies demonstrate that both orthologs are widely expressed with strong presence in embryonic tissues affected in PPS. In vitro glucosylation assays demonstrated that extracts from wildtype embryos contain active b3glct enzyme capable of transferring glucose from UDP-glucose to an O-fucosylated TSR, indicating functional conservation with human B3GLCT. To determine the developmental role of the zebrafish genes, single and double b3glct knockouts were generated using TALEN-induced genome editing. Extracts from double homozygous b3glct-/- embryos demonstrated complete loss of in vitro b3glct activity. Surprisingly, b3glct-/- homozygous fish developed normally. Transcriptome analyses of head and trunk tissues of b3glct-/- 24-hpf embryos identified 483 shared differentially regulated transcripts that may be involved in compensation for b3glct function in these embryos. The presented data show that both sequence and function of B3GLCT/b3glct genes is conserved in vertebrates. At the same time, complete b3glct deficiency in zebrafish appears to be inconsequential and possibly compensated for by a yet unknown mechanism.
Collapse
|
23
|
Charoy C, Dinvaut S, Chaix Y, Morlé L, Sanyas I, Bozon M, Kindbeiter K, Durand B, Skidmore JM, De Groef L, Seki M, Moons L, Ruhrberg C, Martin JF, Martin DM, Falk J, Castellani V. Genetic specification of left-right asymmetry in the diaphragm muscles and their motor innervation. eLife 2017. [PMID: 28639940 PMCID: PMC5481184 DOI: 10.7554/elife.18481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The diaphragm muscle is essential for breathing in mammals. Its asymmetric elevation during contraction correlates with morphological features suggestive of inherent left–right (L/R) asymmetry. Whether this asymmetry is due to L versus R differences in the muscle or in the phrenic nerve activity is unknown. Here, we have combined the analysis of genetically modified mouse models with transcriptomic analysis to show that both the diaphragm muscle and phrenic nerves have asymmetries, which can be established independently of each other during early embryogenesis in pathway instructed by Nodal, a morphogen that also conveys asymmetry in other organs. We further found that phrenic motoneurons receive an early L/R genetic imprint, with L versus R differences both in Slit/Robo signaling and MMP2 activity and in the contribution of both pathways to establish phrenic nerve asymmetry. Our study therefore demonstrates L–R imprinting of spinal motoneurons and describes how L/R modulation of axon guidance signaling helps to match neural circuit formation to organ asymmetry. DOI:http://dx.doi.org/10.7554/eLife.18481.001 The diaphragm is a dome-shaped muscle that forms the floor of the rib cage, separating the lungs from the abdomen. As we breathe in, the diaphragm contracts. This causes the chest cavity to expand, drawing air into the lungs. A pair of nerves called the phrenic nerves carry signals from the spinal cord to the diaphragm to tell it when to contract. These nerves project from the left and right sides of the spinal cord to the left and right sides of the diaphragm respectively. The left and right sides of the diaphragm are not entirely level, but it was not known why. To investigate, Charoy et al. studied how the diaphragm develops in mouse embryos. This revealed that the left and right phrenic nerves are not symmetrical. Neither are the muscles on each side of the diaphragm. Further investigation revealed that a genetic program that establishes other differences between the left and right sides of the embryo also gives rise to the differences between the left and right sides of the diaphragm. This program switches on different genes in the left and right phrenic nerves, which activate different molecular pathways in the left and right sides of the diaphragm muscle. The differences between the nerves and muscles on the left and right sides of the diaphragm could explain why some muscle disorders affect only one side of the diaphragm. Similarly, they could explain why congenital hernias caused by abdominal organs pushing through the diaphragm into the chest cavity mostly affect the left side of the diaphragm. Further studies are now needed to investigate these possibilities. The techniques used by Charoy et al. to map the molecular diversity of spinal cord neurons could also lead to new strategies for repairing damage to the spinal cord following injury or disease. DOI:http://dx.doi.org/10.7554/eLife.18481.002
Collapse
Affiliation(s)
- Camille Charoy
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Sarah Dinvaut
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Yohan Chaix
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Laurette Morlé
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Isabelle Sanyas
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Muriel Bozon
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Karine Kindbeiter
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Bénédicte Durand
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Jennifer M Skidmore
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, United States.,Department of Communicable Diseases, University of Michigan Medical Center, Ann Arbor, United States
| | - Lies De Groef
- Animal Physiology and Neurobiology Section, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Leuven, Belgium
| | - Motoaki Seki
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Lieve Moons
- Animal Physiology and Neurobiology Section, Department of Biology, Laboratory of Neural Circuit Development and Regeneration, Leuven, Belgium
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | - Donna M Martin
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, United States.,Department of Communicable Diseases, University of Michigan Medical Center, Ann Arbor, United States.,Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, United States
| | - Julien Falk
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| | - Valerie Castellani
- University of Lyon, Claude Bernard University Lyon 1, INMG UMR CNRS 5310, INSERM U1217, Lyon, France
| |
Collapse
|
24
|
Gao Q, Zhang J, Wang X, Liu Y, He R, Liu X, Wang F, Feng J, Yang D, Wang Z, Meng A, Yan X. The signalling receptor MCAM coordinates apical-basal polarity and planar cell polarity during morphogenesis. Nat Commun 2017; 8:15279. [PMID: 28589943 PMCID: PMC5467231 DOI: 10.1038/ncomms15279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
The apical-basal (AB) polarity and planar cell polarity (PCP) provide an animal cell population with different phenotypes during morphogenesis. However, how cells couple these two patterning systems remains unclear. Here we provide in vivo evidence that melanoma cell adhesion molecule (MCAM) coordinates AB polarity-driven lumenogenesis and c-Jun N-terminal kinase (JNK)/PCP-dependent ciliogenesis. We identify that MCAM is an independent receptor of fibroblast growth factor 4 (FGF4), a membrane anchor of phospholipase C-γ (PLC-γ), an immediate upstream receptor of nuclear factor of activated T-cells (NFAT) and a constitutive activator of JNK. We find that MCAM-mediated vesicular trafficking towards FGF4, while generating a priority-grade transcriptional response of NFAT determines lumenogenesis. We demonstrate that MCAM plays indispensable roles in ciliogenesis through activating JNK independently of FGF signals. Furthermore, mcam-deficient zebrafish and Xenopus exhibit a global defect in left-right (LR) asymmetric establishment as a result of morphogenetic failure of their LR organizers. Therefore, MCAM coordination of AB polarity and PCP provides insight into the general mechanisms of morphogenesis.
Collapse
Affiliation(s)
- Qian Gao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junfeng Zhang
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiumei Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rongqiao He
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingfeng Liu
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fei Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Feng
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongling Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Anming Meng
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
25
|
Pitcairn E, Harris H, Epiney J, Pai VP, Lemire JM, Ye B, Shi NQ, Levin M, McLaughlin KA. Coordinating heart morphogenesis: A novel role for hyperpolarization-activated cyclic nucleotide-gated (HCN) channels during cardiogenesis in Xenopus laevis. Commun Integr Biol 2017; 10:e1309488. [PMID: 28702127 PMCID: PMC5501196 DOI: 10.1080/19420889.2017.1309488] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
Hyperpolarization-activated cyclic-nucleotide gated channel (HCN) proteins are important regulators of both neuronal and cardiac excitability. Among the 4 HCN isoforms, HCN4 is known as a pacemaker channel, because it helps control the periodicity of contractions in vertebrate hearts. Although the physiological role of HCN4 channel has been studied in adult mammalian hearts, an earlier role during embryogenesis has not been clearly established. Here, we probe the embryonic roles of HCN4 channels, providing the first characterization of the expression profile of any of the HCN isoforms during Xenopus laevis development and investigate the consequences of altering HCN4 function on embryonic pattern formation. We demonstrate that both overexpression of HCN4 and injection of dominant-negative HCN4 mRNA during early embryogenesis results in improper expression of key patterning genes and severely malformed hearts. Our results suggest that HCN4 serves to coordinate morphogenetic control factors that provide positional information during heart morphogenesis in Xenopus.
Collapse
Affiliation(s)
- Emily Pitcairn
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Hannah Harris
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Justine Epiney
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Vaibhav P Pai
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Joan M Lemire
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Bin Ye
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nian-Qing Shi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Levin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Kelly A McLaughlin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| |
Collapse
|
26
|
Duboué ER, Halpern ME. Genetic and Transgenic Approaches to Study Zebrafish Brain Asymmetry and Lateralized Behavior. LATERALIZED BRAIN FUNCTIONS 2017. [DOI: 10.1007/978-1-4939-6725-4_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|
27
|
Reade A, Motta-Mena LB, Gardner KH, Stainier DY, Weiner OD, Woo S. TAEL: a zebrafish-optimized optogenetic gene expression system with fine spatial and temporal control. Development 2016; 144:345-355. [PMID: 27993986 DOI: 10.1242/dev.139238] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 11/28/2016] [Indexed: 01/27/2023]
Abstract
Here, we describe an optogenetic gene expression system optimized for use in zebrafish. This system overcomes the limitations of current inducible expression systems by enabling robust spatial and temporal regulation of gene expression in living organisms. Because existing optogenetic systems show toxicity in zebrafish, we re-engineered the blue-light-activated EL222 system for minimal toxicity while exhibiting a large range of induction, fine spatial precision and rapid kinetics. We validate several strategies to spatially restrict illumination and thus gene induction with our new TAEL (TA4-EL222) system. As a functional example, we show that TAEL is able to induce ectopic endodermal cells in the presumptive ectoderm via targeted sox32 induction. We also demonstrate that TAEL can be used to resolve multiple roles of Nodal signaling at different stages of embryonic development. Finally, we show how inducible gene editing can be achieved by combining the TAEL and CRISPR/Cas9 systems. This toolkit should be a broadly useful resource for the fish community.
Collapse
Affiliation(s)
- Anna Reade
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Laura B Motta-Mena
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, NY 10031, USA
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, NY 10031, USA.,Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031, USA.,Biochemistry, Chemistry and Biology PhD Programs, Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Didier Y Stainier
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.,Max Planck Institute for Heart and Lung Research, Dept. of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Orion D Weiner
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephanie Woo
- CVRI & Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
28
|
Zhang X, Zhang S, Yang Q, Lei C, Chen H, Lan X. Exploration of dairy goat PITX2 alternative splice events and differential isoform expression. Small Rumin Res 2016. [DOI: 10.1016/j.smallrumres.2016.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
29
|
Ji Y, Buel SM, Amack JD. Mutations in zebrafish pitx2 model congenital malformations in Axenfeld-Rieger syndrome but do not disrupt left-right placement of visceral organs. Dev Biol 2016; 416:69-81. [PMID: 27297886 DOI: 10.1016/j.ydbio.2016.06.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 06/05/2016] [Accepted: 06/06/2016] [Indexed: 12/18/2022]
Abstract
Pitx2 is a conserved homeodomain transcription factor that has multiple functions during embryonic development. Mutations in human PITX2 cause autosomal dominant Axenfeld-Rieger syndrome (ARS), characterized by congenital eye and tooth malformations. Pitx2(-/-) knockout mouse models recapitulate aspects of ARS, but are embryonic lethal. To date, ARS treatments remain limited to managing individual symptoms due to an incomplete understanding of PITX2 function. In addition to regulating eye and tooth development, Pitx2 is a target of a conserved Nodal (TGFβ) signaling pathway that mediates left-right (LR) asymmetry of visceral organs. Based on its highly conserved asymmetric expression domain, the Nodal-Pitx2 axis has long been considered a common denominator of LR development in vertebrate embryos. However, functions of Pitx2 during asymmetric organ morphogenesis are not well understood. To gain new insight into Pitx2 function we used genome editing to create mutations in the zebrafish pitx2 gene. Mutations in the pitx2 homeodomain caused phenotypes reminiscent of ARS, including aberrant development of the cornea and anterior chamber of the eye and reduced or absent teeth. Intriguingly, LR asymmetric looping of the heart and gut was normal in pitx2 mutants. These results suggest conserved roles for Pitx2 in eye and tooth development and indicate Pitx2 is not required for asymmetric looping of zebrafish visceral organs. This work establishes zebrafish pitx2 mutants as a new animal model for investigating mechanisms underlying congenital malformations in ARS and high-throughput drug screening for ARS therapeutics. Additionally, pitx2 mutants present a unique opportunity to identify new genes involved in vertebrate LR patterning. We show Nodal signaling-independent of Pitx2-controls asymmetric expression of the fatty acid elongase elovl6 in zebrafish, pointing to a potential novel pathway during LR organogenesis.
Collapse
Affiliation(s)
- Yongchang Ji
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Sharleen M Buel
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
30
|
Nagelberg D, Wang J, Su R, Torres-Vázquez J, Targoff KL, Poss KD, Knaut H. Origin, Specification, and Plasticity of the Great Vessels of the Heart. Curr Biol 2015; 25:2099-110. [PMID: 26255850 PMCID: PMC4546555 DOI: 10.1016/j.cub.2015.06.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/02/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
Abstract
The pharyngeal arch arteries (PAAs) are a series of paired embryonic blood vessels that give rise to several major arteries that connect directly to the heart. During development, the PAAs emerge from nkx2.5-expressing mesodermal cells and connect the dorsal head vasculature to the outflow tract of the heart. Despite their central role in establishing the circulatory system, the embryonic origins of the PAA progenitors are only coarsely defined, and the factors that specify them and their regenerative potential are unclear. Using fate mapping and mutant analysis, we find that PAA progenitors are derived from the tcf21 and nkx2.5 double-positive head mesoderm and require these two transcription factors for their specification and survival. Unexpectedly, cell ablation shows that the tcf21+; nkx2.5+ PAA progenitors are not required for PAA formation. We find that this compensation is due to the replacement of ablated tcf21+; nkx2.5+ PAA cells by endothelial cells from the dorsal head vasculature. Together, these studies assign the embryonic origin of the great vessel progenitors to the interface between the pharyngeal and cardiac mesoderm, identify the transcription factor code required for their specification, and reveal an unexpected plasticity in the formation of the great vessels.
Collapse
Affiliation(s)
- Danielle Nagelberg
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jinhu Wang
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, 349 Nanaline Duke Building, Durham, NC 27710, USA
| | - Rina Su
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jesús Torres-Vázquez
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Kimara L Targoff
- College of Physicians and Surgeons, Columbia University Medical Center, 630 West 168(th) Street, New York, NY 10023, USA
| | - Kenneth D Poss
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, 349 Nanaline Duke Building, Durham, NC 27710, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
31
|
Casar Tena T, Burkhalter MD, Philipp M. Left-right asymmetry in the light of TOR: An update on what we know so far. Biol Cell 2015; 107:306-18. [PMID: 25943139 PMCID: PMC4744706 DOI: 10.1111/boc.201400094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/29/2015] [Indexed: 01/06/2023]
Abstract
The internal left‐right (LR) asymmetry is a characteristic that exists throughout the animal kingdom from roundworms over flies and fish to mammals. Cilia, which are antenna‐like structures protruding into the extracellular space, are involved in establishing LR asymmetry during early development. Humans who suffer from dysfunctional cilia often develop conditions such as heterotaxy, where internal organs appear to be placed randomly. As a consequence to this failure in asymmetry development, serious complications such as congenital heart defects (CHD) occur. The mammalian (or mechanistic) target of rapamycin (mTOR) pathway has recently emerged as an important regulator regarding symmetry breaking. The mTOR pathway governs fundamental processes such as protein translation or metabolism. Its activity can be transduced by two complexes, which are called TORC1 and TORC2, respectively. So far, only TORC1 has been implicated with asymmetry development and appears to require very precise regulation. A number of recent papers provided evidence that dysregulated TORC1 results in alterations of motile cilia and asymmetry defects. In here, we give an update on what we know so far of mTORC1 in LR asymmetry development.
Collapse
Affiliation(s)
- Teresa Casar Tena
- Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| | - Martin D Burkhalter
- Leibniz Institute for Age Research Fritz Lippmann Institute, Jena, 07745, Germany
| | - Melanie Philipp
- Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, 89081, Germany
| |
Collapse
|
32
|
LeftyA sensitive cytosolic pH regulation and glycolytic flux in Ishikawa human endometrial cancer cells. Biochem Biophys Res Commun 2015; 460:845-9. [PMID: 25838200 DOI: 10.1016/j.bbrc.2015.03.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE LeftyA, a powerful regulator of stemness, embryonic differentiation, and reprogramming of cancer cells, counteracts cell proliferation and tumor growth. Key properties of tumor cells include enhanced glycolytic flux, which is highly sensitive to cytosolic pH and thus requires export of H(+) and lactate. H(+) extrusion is in part accomplished by Na(+)/H(+) exchangers, such as NHE1. An effect of LeftyA on transport processes has, however, never been reported. The present study thus explored whether LeftyA modifies regulation of cytosolic pH (pHi) in Ishikawa cells, a well differentiated endometrial carcinoma cell model. METHODS NHE1 transcript levels were determined by qRT-PCR, NHE1 protein abundance quantified by Western blotting, pHi estimated utilizing (2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein [BCECF] fluorescence, Na(+)/H(+) exchanger activity from Na(+) dependent realkalinization after an ammonium pulse, and lactate concentration in the supernatant utilizing an enzymatic assay and subsequent colorimetry. RESULTS A 2 h treatment with LeftyA (8 ng/ml) significantly decreased NHE1 transcript levels (by 99.6%), NHE1 protein abundance (by 71%), Na(+)/H(+) exchanger activity (by 55%), pHi (from 7.22 ± 0.02 to 7.05 ± 0.02), and lactate release (by 41%). CONCLUSIONS LeftyA markedly down-regulates NHE1 expression, Na(+)/H(+) exchanger activity, pHi, and lactate release in Ishikawa cells. Those effects presumably contribute to cellular reprogramming and growth inhibition.
Collapse
|
33
|
Doucette LP, Rasnitsyn A, Seifi M, Walter MA. The interactions of genes, age, and environment in glaucoma pathogenesis. Surv Ophthalmol 2015; 60:310-26. [PMID: 25907525 DOI: 10.1016/j.survophthal.2015.01.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/27/2015] [Accepted: 01/29/2015] [Indexed: 12/30/2022]
Abstract
Glaucoma, a progressive degenerative condition that results in the death of retinal ganglion cells, is one of the leading causes of blindness, affecting millions worldwide. The mechanisms underlying glaucoma are not well understood, although years of studies have shown that the largest risk factors are elevated intraocular pressure, age, and genetics. Eleven genes and multiple loci have been identified as contributing factors. These genes act by a number of mechanisms, including mechanical stress, ischemic/oxidative stress, and neurodegeneration. We summarize the recent advances in the understanding of glaucoma and propose a unified hypothesis for glaucoma pathogenesis. Glaucoma does not result from a single pathological mechanism, but rather a combination of pathways that are influenced by genes, age, and environment. In particular, we hypothesize that, in the presence of genetic risk factors, exposure to environment stresses results in an earlier age of onset for glaucoma. This hypothesis is based upon the overlap of the molecular pathways in which glaucoma genes are involved. Because of the interactions between these processes, it is likely that there are common therapies that may be effective for different subtypes of glaucoma.
Collapse
Affiliation(s)
- Lance P Doucette
- Faculty of Medicine and Dentistry, Department of Medical Genetics, Edmonton, Alberta T6G 2H7, Canada
| | - Alexandra Rasnitsyn
- Faculty of Medicine and Dentistry, Department of Medical Genetics, Edmonton, Alberta T6G 2H7, Canada
| | - Morteza Seifi
- Faculty of Medicine and Dentistry, Department of Medical Genetics, Edmonton, Alberta T6G 2H7, Canada
| | - Michael A Walter
- Faculty of Medicine and Dentistry, Department of Medical Genetics, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
34
|
Waite MR, Martin DM. Axial level-specific regulation of neuronal development: lessons from PITX2. J Neurosci Res 2015; 93:195-8. [PMID: 25124216 DOI: 10.1002/jnr.23471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/09/2014] [Accepted: 07/16/2014] [Indexed: 12/19/2022]
Abstract
Transcriptional regulation of gene expression is vital for proper control of proliferation, migration, differentiation, and survival of developing neurons. Pitx2 encodes a homeodomain transcription factor that is highly expressed in the developing and adult mammalian brain. In humans, mutations in PITX2 result in Rieger syndrome, characterized by defects in the development of the eyes, umbilicus, and teeth and variable abnormalities in the brain, including hydrocephalus and cerebellar hypoplasia. Alternative splicing of Pitx2 in the mouse results in three isoforms, Pitx2a, Pitx2b, and Pitx2c, each of which is expressed symmetrically along the left-right axis of the brain throughout development. Here, we review recent evidence for axial and brain region-specific requirements for Pitx2 during neuronal migration and differentiation, highlighting known isoform contributions.
Collapse
Affiliation(s)
- Mindy R Waite
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
35
|
Alexander C, Piloto S, Le Pabic P, Schilling TF. Wnt signaling interacts with bmp and edn1 to regulate dorsal-ventral patterning and growth of the craniofacial skeleton. PLoS Genet 2014; 10:e1004479. [PMID: 25058015 PMCID: PMC4109847 DOI: 10.1371/journal.pgen.1004479] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 05/16/2014] [Indexed: 11/25/2022] Open
Abstract
Craniofacial development requires signals from epithelia to pattern skeletogenic neural crest (NC) cells, such as the subdivision of each pharyngeal arch into distinct dorsal (D) and ventral (V) elements. Wnt signaling has been implicated in many aspects of NC and craniofacial development, but its roles in D-V arch patterning remain unclear. To address this we blocked Wnt signaling in zebrafish embryos in a temporally-controlled manner, using transgenics to overexpress a dominant negative Tcf3, (dntcf3), (Tg(hsp70I:tcf3-GFP), or the canonical Wnt inhibitor dickkopf1 (dkk1), (Tg(hsp70i:dkk1-GFP) after NC migration. In dntcf3 transgenics, NC cells in the ventral arches of heat-shocked embryos show reduced proliferation, expression of ventral patterning genes (hand2, dlx3b, dlx5a, msxe), and ventral cartilage differentiation (e.g. lower jaws). These D-V patterning defects resemble the phenotypes of zebrafish embryos lacking Bmp or Edn1 signaling, and overexpression of dntcf3 dramatically reduces expression of a subset of Bmp receptors in the arches. Addition of ectopic BMP (or EDN1) protein partially rescues ventral development and expression of dlx3b, dlx5a, and msxe in Wnt signaling-deficient embryos, but surprisingly does not rescue hand2 expression. Thus Wnt signaling provides ventralizing patterning cues to arch NC cells, in part through regulation of Bmp and Edn1 signaling, but independently regulates hand2. Similarly, heat-shocked dkk1+ embryos exhibit ventral arch reductions, but also have mandibular clefts at the ventral midline not seen in dntcf3+ embryos. Dkk1 is expressed in pharyngeal endoderm, and cell transplantation experiments reveal that dntcf3 must be overexpressed in pharyngeal endoderm to disrupt D-V arch patterning, suggesting that distinct endodermal roles for Wnts and Wnt antagonists pattern the developing skeleton. Craniofacial abnormalities are among the most common birth defects. Understanding the molecular mechanisms underlying craniofacial disorders is crucial for developing treatment strategies. Much of the craniofacial skeleton arises from specialized embryonic structures known as pharyngeal arches. Patterning of these arches requires precise spatial and temporal expression of multiple genes, which is coordinated between tissues by secreted signals. Wnts are secreted ligands expressed throughout the pharyngeal arches yet their role in craniofacial patterning remains unclear. In this study we examine the role of Wnts in craniofacial patterning using transgenic zebrafish to inhibit downstream Wnt signaling. We show that Wnt signaling deficient embryos have lower jaw specific defects, which strongly resembles loss-of-function phenotypes in both the Bmp and Edn1 signaling pathways. Through rescue experiments we find that Wnts are upstream regulators of both Bmp and Edn1 signaling. We thus have uncovered a crucial requirement for Wnt signaling in craniofacial patterning.
Collapse
Affiliation(s)
- Courtney Alexander
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Sarah Piloto
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Pierre Le Pabic
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Araya C, Tawk M, Girdler GC, Costa M, Carmona-Fontaine C, Clarke JD. Mesoderm is required for coordinated cell movements within zebrafish neural plate in vivo. Neural Dev 2014; 9:9. [PMID: 24755297 PMCID: PMC4022452 DOI: 10.1186/1749-8104-9-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 04/01/2014] [Indexed: 01/24/2023] Open
Abstract
Background Morphogenesis of the zebrafish neural tube requires the coordinated movement of many cells in both time and space. A good example of this is the movement of the cells in the zebrafish neural plate as they converge towards the dorsal midline before internalizing to form a neural keel. How these cells are regulated to ensure that they move together as a coherent tissue is unknown. Previous work in other systems has suggested that the underlying mesoderm may play a role in this process but this has not been shown directly in vivo. Results Here we analyze the roles of subjacent mesoderm in the coordination of neural cell movements during convergence of the zebrafish neural plate and neural keel formation. Live imaging demonstrates that the normal highly coordinated movements of neural plate cells are lost in the absence of underlying mesoderm and the movements of internalization and neural tube formation are severely disrupted. Despite this, neuroepithelial polarity develops in the abnormal neural primordium but the resulting tissue architecture is very disorganized. Conclusions We show that the movements of cells in the zebrafish neural plate are highly coordinated during the convergence and internalization movements of neurulation. Our results demonstrate that the underlying mesoderm is required for these coordinated cell movements in the zebrafish neural plate in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Jonathan Dw Clarke
- Medical Research Council (MRC) Centre for Developmental Neurobiology, King's College London, New Hunt's House, 4th Floor, Guy's Hospital Campus, London SE1 1UL, UK.
| |
Collapse
|
37
|
Garric L, Ronsin B, Roussigné M, Booton S, Gamse JT, Dufourcq P, Blader P. Pitx2c ensures habenular asymmetry by restricting parapineal cell number. Development 2014; 141:1572-9. [PMID: 24598158 DOI: 10.1242/dev.100305] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Left-right (L/R) asymmetries in the brain are thought to underlie lateralised cognitive functions. Understanding how neuroanatomical asymmetries are established has been achieved through the study of the zebrafish epithalamus. Morphological symmetry in the epithalamus is broken by leftward migration of the parapineal, which is required for the subsequent elaboration of left habenular identity; the habenular nuclei flank the midline and show L/R asymmetries in marker expression and connectivity. The Nodal target pitx2c is expressed in the left epithalamus, but nothing is known about its role during the establishment of asymmetry in the brain. We show that abrogating Pitx2c function leads to the right habenula adopting aspects of left character, and to an increase in parapineal cell numbers. Parapineal ablation in Pitx2c loss of function results in right habenular isomerism, indicating that the parapineal is required for the left character detected in the right habenula in this context. Partial parapineal ablation in the absence of Pitx2c, however, reduces the number of parapineal cells to wild-type levels and restores habenular asymmetry. We provide evidence suggesting that antagonism between Nodal and Pitx2c activities sets an upper limit on parapineal cell numbers. We conclude that restricting parapineal cell number is crucial for the correct elaboration of epithalamic asymmetry.
Collapse
Affiliation(s)
- Laurence Garric
- Université de Toulouse, UPS, Centre de Biologie du Développement (CBD), 118 route de Narbonne, F-31062 Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
38
|
A potential molecular pathogenesis of cardiac/laterality defects in Oculo-Facio-Cardio-Dental syndrome. Dev Biol 2014; 387:28-36. [PMID: 24440151 DOI: 10.1016/j.ydbio.2014.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 01/04/2014] [Accepted: 01/09/2014] [Indexed: 01/16/2023]
Abstract
Pitx2 is the last effector of the left-right (LR) cascade known to date and plays a crucial role in the patterning of LR asymmetry. In Xenopus embryos, the expression of Pitx2 gene in the left lateral plate mesoderm (LPM) is directly regulated by Xnr1 signaling, which is mediated by Smads and FoxH1. Previous studies suggest that the suppression of Pitx2 gene in the left LPM is a potential cause of cardiac/laterality defects in Oculo-Facio-Cardio-Dental (OFCD) syndrome, which is known to be caused by mutations in BCL6 co-repressor (BCOR) gene. Recently, our work has revealed that the BCL6/BCOR complex blocks Notch-dependent transcriptional activity to protect the expression of Pitx2 in the left LPM from the inhibitory activity of Notch signaling. These studies indicated that uncontrolled Notch activity in the left LPM caused by dysfunction of BCOR may result in cardiac/laterality defects of OFCD syndrome. However, this Notch-dependent inhibitory mechanism of Pitx2 gene transcription still remains unknown. Here we report that transcriptional repressor ESR1, which acts downstream of Notch signaling, inhibits the expression of Pitx2 gene by binding to a left side-specific enhancer (ASE) region in Pitx2 gene and recruiting histone deacetylase 1 (HDAC1) to this region. Once HDAC1 is tethered, histone acetyltransferase p300 is no longer recruited to the Xnr1-dependent transcriptional complex on the ASE region, leading to the suppression of Pitx2 gene in the left LPM. The study presented here uncovers the regulatory mechanism of Pitx2 gene transcription which may contribute to an understanding of pathogenesis of OFCD syndrome.
Collapse
|
39
|
Samson SC, Ferrer T, Jou CJ, Sachse FB, Shankaran SS, Shaw RM, Chi NC, Tristani-Firouzi M, Yost HJ. 3-OST-7 regulates BMP-dependent cardiac contraction. PLoS Biol 2013; 11:e1001727. [PMID: 24311987 PMCID: PMC3849020 DOI: 10.1371/journal.pbio.1001727] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/23/2013] [Indexed: 11/29/2022] Open
Abstract
During zebrafish cardiac development, 3-OST-7 constrains BMP signaling to the atrioventricular junction and precludes it from contractile myocardium, allowing tropomyosin-dependent sarcomere assembly and contraction. The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function. A highly complex environment at the cell surface and in the space between cells is thought to modulate cell behavior. Heparan sulfate proteoglycans are cell surface and extracellular matrix molecules that are covalently linked to long chains of repeating sugar units called glycosaminoglycan chains. These chains can be subjected to rare modifications and they are believed to influence specific cell signaling events in a lineage specific fashion in what is called the “glycocode.” Here we explore the functions of one member of a family of enzymes, 3-O-sulfotransferases (3-OSTs) that catalyzes a rare modification (3-O-sulfation) of glycosaminoglycans in zebrafish. We show that knockdown of 3-OST-7 results in a very specific phenotype, including loss of cardiac ventricle contraction. Knockdown of other 3-OST family members did not result in the same phenotype, suggesting that distinct 3-OST family members have distinct functions in vertebrates and lending in vivo evidence for the glycocode hypothesis. Mechanistically, we found that cardiac contraction can be rescued by reducing the amount of endogenous BMP4, and can be blocked by increasing BMP signaling, suggesting that the glycocode generated by 3-OST-7 is necessary to constrain BMP signaling in the heart for normal cardiac contraction. Furthermore, we show that tropomyosin4 (tpm4) is downstream of 3-OST-7 function, indicating that Tpm4 is key in this pathway to building the sarcomere, the functional contraction unit of the cardiomyocyte.
Collapse
Affiliation(s)
- Shiela C. Samson
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
| | - Tania Ferrer
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Chuanchau J. Jou
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Frank B. Sachse
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Sunita S. Shankaran
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Robin M. Shaw
- Department of Medicine, Cedars-Sinai Heart Institute, University of California, Los Angeles, California, United States of America
| | - Neil C. Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, California, United States of America
| | - Martin Tristani-Firouzi
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - H. Joseph Yost
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
40
|
Colombo A, Palma K, Armijo L, Mione M, Signore IA, Morales C, Guerrero N, Meynard MM, Pérez R, Suazo J, Marcelain K, Briones L, Härtel S, Wilson SW, Concha ML. Daam1a mediates asymmetric habenular morphogenesis by regulating dendritic and axonal outgrowth. Development 2013; 140:3997-4007. [PMID: 24046318 PMCID: PMC3775416 DOI: 10.1242/dev.091934] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Although progress has been made in resolving the genetic pathways that specify neuronal asymmetries in the brain, little is known about genes that mediate the development of structural asymmetries between neurons on left and right. In this study, we identify daam1a as an asymmetric component of the signalling pathways leading to asymmetric morphogenesis of the habenulae in zebrafish. Daam1a is a member of the Formin family of actin-binding proteins and the extent of Daam1a expression in habenular neuron dendrites mirrors the asymmetric growth of habenular neuropil between left and right. Local loss and gain of Daam1a function affects neither cell number nor subtype organisation but leads to a decrease or increase of neuropil, respectively. Daam1a therefore plays a key role in the asymmetric growth of habenular neuropil downstream of the pathways that specify asymmetric cellular domains in the habenulae. In addition, Daam1a mediates the development of habenular efferent connectivity as local loss and gain of Daam1a function impairs or enhances, respectively, the growth of habenular neuron terminals in the interpeduncular nucleus. Abrogation of Daam1a disrupts the growth of both dendritic and axonal processes and results in disorganised filamentous actin and α-tubulin. Our results indicate that Daam1a plays a key role in asymmetric habenular morphogenesis mediating the growth of dendritic and axonal processes in dorsal habenular neurons.
Collapse
Affiliation(s)
- Alicia Colombo
- Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Small heat shock proteins are necessary for heart migration and laterality determination in zebrafish. Dev Biol 2013; 384:166-80. [PMID: 24140541 DOI: 10.1016/j.ydbio.2013.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/27/2022]
Abstract
Small heat shock proteins (sHsps) regulate cellular functions not only under stress, but also during normal development, when they are expressed in organ-specific patterns. Here we demonstrate that two small heat shock proteins expressed in embryonic zebrafish heart, hspb7 and hspb12, have roles in the development of left-right asymmetry. In zebrafish, laterality is determined by the motility of cilia in Kupffer's vesicle (KV), where hspb7 is expressed; knockdown of hspb7 causes laterality defects by disrupting the motility of these cilia. In embryos with reduced hspb7, the axonemes of KV cilia have a 9+0 structure, while control embyros have a predominately 9+2 structure. Reduction of either hspb7 or hspb12 alters the expression pattern of genes that propagate the signals that establish left-right asymmetry: the nodal-related gene southpaw (spaw) in the lateral plate mesoderm, and its downstream targets pitx2, lefty1 and lefty2. Partial depletion of hspb7 causes concordant heart, brain and visceral laterality defects, indicating that loss of KV cilia motility leads to coordinated but randomized laterality. Reducing hspb12 leads to similar alterations in the expression of downstream laterality genes, but at a lower penetrance. Simultaneous reduction of hspb7 and hspb12 randomizes heart, brain and visceral laterality, suggesting that these two genes have partially redundant functions in the establishment of left-right asymmetry. In addition, both hspb7 and hspb12 are expressed in the precardiac mesoderm and in the yolk syncytial layer, which supports the migration and fusion of mesodermal cardiac precursors. In embryos in which the reduction of hspb7 or hspb12 was limited to the yolk, migration defects predominated, suggesting that the yolk expression of these genes rather than heart expression is responsible for the migration defects.
Collapse
|
42
|
Hochgreb-Hägele T, Yin C, Koo DES, Bronner ME, Stainier DYR. Laminin β1a controls distinct steps during the establishment of digestive organ laterality. Development 2013; 140:2734-45. [PMID: 23757411 DOI: 10.1242/dev.097618] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Visceral organs, including the liver and pancreas, adopt asymmetric positions to ensure proper function. Yet the molecular and cellular mechanisms controlling organ laterality are not well understood. We identified a mutation affecting zebrafish laminin β1a (lamb1a) that disrupts left-right asymmetry of the liver and pancreas. In these mutants, the liver spans the midline and the ventral pancreatic bud remains split into bilateral structures. We show that lamb1a regulates asymmetric left-right gene expression in the lateral plate mesoderm (LPM). In particular, lamb1a functions in Kupffer's vesicle (KV), a ciliated organ analogous to the mouse node, to control the length and function of the KV cilia. Later during gut-looping stages, dynamic expression of Lamb1a is required for the bilayered organization and asymmetric migration of the LPM. Loss of Lamb1a function also results in aberrant protrusion of LPM cells into the gut. Collectively, our results provide cellular and molecular mechanisms by which extracellular matrix proteins regulate left-right organ morphogenesis.
Collapse
Affiliation(s)
- Tatiana Hochgreb-Hägele
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Liver Center and Diabetes Center, Institute for Regeneration Medicine, University of California, San Francisco, CA 94158, USA.
| | | | | | | | | |
Collapse
|
43
|
Neugebauer JM, Cadwallader AB, Amack JD, Bisgrove BW, Yost HJ. Differential roles for 3-OSTs in the regulation of cilia length and motility. Development 2013; 140:3892-902. [PMID: 23946439 DOI: 10.1242/dev.096388] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
As cells integrate molecular signals from their environment, cell surface receptors require modified proteoglycans for the robust activation of signaling pathways. Heparan sulfate proteoglycans (HSPGs) have long unbranched chains of repetitive disaccharide units that can be sulfated at specific positions by heparan sulfate O-sulfotransferase (OST) families. Here, we show that two members of the 3-OST family are required in distinct signaling pathways to control left-right (LR) patterning through control of Kupffer's vesicle (KV) cilia length and motility. 3-OST-5 functions in the fibroblast growth factor pathway to control cilia length via the ciliogenic transcription factors FoxJ1a and Rfx2. By contrast, a second 3-OST family member, 3-OST-6, does not regulate cilia length, but regulates cilia motility via kinesin motor molecule (Kif3b) expression and cilia arm dynein assembly. Thus, two 3-OST family members cell-autonomously control LR patterning through distinct pathways that regulate KV fluid flow. We propose that individual 3-OST isozymes create distinct modified domains or 'glycocodes' on cell surface proteoglycans, which in turn regulate the response to diverse cell signaling pathways.
Collapse
Affiliation(s)
- Judith M Neugebauer
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
44
|
Tay HG, Schulze SK, Compagnon J, Foley FC, Heisenberg CP, Yost HJ, Abdelilah-Seyfried S, Amack JD. Lethal giant larvae 2 regulates development of the ciliated organ Kupffer's vesicle. Development 2013; 140:1550-9. [PMID: 23482490 DOI: 10.1242/dev.087130] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Motile cilia perform crucial functions during embryonic development and throughout adult life. Development of organs containing motile cilia involves regulation of cilia formation (ciliogenesis) and formation of a luminal space (lumenogenesis) in which cilia generate fluid flows. Control of ciliogenesis and lumenogenesis is not yet fully understood, and it remains unclear whether these processes are coupled. In the zebrafish embryo, lethal giant larvae 2 (lgl2) is expressed prominently in ciliated organs. Lgl proteins are involved in establishing cell polarity and have been implicated in vesicle trafficking. Here, we identified a role for Lgl2 in development of ciliated epithelia in Kupffer's vesicle, which directs left-right asymmetry of the embryo; the otic vesicles, which give rise to the inner ear; and the pronephric ducts of the kidney. Using Kupffer's vesicle as a model ciliated organ, we found that depletion of Lgl2 disrupted lumen formation and reduced cilia number and length. Immunofluorescence and time-lapse imaging of Kupffer's vesicle morphogenesis in Lgl2-deficient embryos suggested cell adhesion defects and revealed loss of the adherens junction component E-cadherin at lateral membranes. Genetic interaction experiments indicate that Lgl2 interacts with Rab11a to regulate E-cadherin and mediate lumen formation that is uncoupled from cilia formation. These results uncover new roles and interactions for Lgl2 that are crucial for both lumenogenesis and ciliogenesis and indicate that these processes are genetically separable in zebrafish.
Collapse
Affiliation(s)
- Hwee Goon Tay
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Teslaa JJ, Keller AN, Nyholm MK, Grinblat Y. Zebrafish Zic2a and Zic2b regulate neural crest and craniofacial development. Dev Biol 2013; 380:73-86. [PMID: 23665173 DOI: 10.1016/j.ydbio.2013.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 11/25/2022]
Abstract
Holoprosencephaly (HPE), the most common malformation of the human forebrain, is associated with defects of the craniofacial skeleton. ZIC2, a zinc-finger transcription factor, is strongly linked to HPE and to a characteristic set of dysmorphic facial features in humans. We have previously identified important functions for zebrafish Zic2 in the developing forebrain. Here, we demonstrate that ZIC2 orthologs zic2a and zic2b also regulate the forming zebrafish craniofacial skeleton, including the jaw and neurocranial cartilages, and use the zebrafish to study Zic2-regulated processes that may contribute to the complex etiology of HPE. Using temporally controlled Zic2a overexpression, we show that the developing craniofacial cartilages are sensitive to Zic2 elevation prior to 24hpf. This window of sensitivity overlaps the critical expansion and migration of the neural crest (NC) cells, which migrate from the developing neural tube to populate vertebrate craniofacial structures. We demonstrate that zic2b influences the induction of NC at the neural plate border, while both zic2a and zic2b regulate NC migratory onset and strongly contribute to chromatophore development. Both Zic2 depletion and early ectopic Zic2 expression cause moderate, incompletely penetrant mispatterning of the NC-derived jaw precursors at 24hpf, yet by 2dpf these changes in Zic2 expression result in profoundly mispatterned chondrogenic condensations. We attribute this discrepancy to an additional role for Zic2a and Zic2b in patterning the forebrain primordium, an important signaling source during craniofacial development. This hypothesis is supported by evidence that transplanted Zic2-deficient cells can contribute to craniofacial cartilages in a wild-type background. Collectively, these data suggest that zebrafish Zic2 plays a dual role during craniofacial development, contributing to two disparate aspects of craniofacial morphogenesis: (1) neural crest induction and migration, and (2) early patterning of tissues adjacent to craniofacial chondrogenic condensations.
Collapse
Affiliation(s)
- Jessica J Teslaa
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
46
|
Smoczer C, Hooker L, Brode S, Wolanski M, KhosrowShahian F, Crawford M. The Xenopus homeobox gene pitx3 impinges upon somitogenesis and laterality. Biochem Cell Biol 2013; 91:79-87. [PMID: 23527636 DOI: 10.1139/bcb-2012-0057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pitx3 has been identified as the causative locus in a developmental eye mutation associated with mammalian anterior segment dysgenesis, congenital cataracts, and aphakia. In recent studies of frog eye development we discovered that pitx3 expresses symmetrically in the somites and lateral plate mesoderm and asymmetrically during cardiac and gut looping. We report that disruption of pitx3 activity on one side of an embryo relative to the other, either by over- or underexpression of pitx3, elicits a crooked dorsal axis in embryos that is a consequence of a retarded progression through somitogenesis. Unlike in amniotes, Xenopus somites form as cohorts of presomitic cells that rotate perpendicular to the dorsal axis. Since no vertebral anomalies have been reported in mouse and human Pitx3 mutants, we attempt to distinguish whether the segmentation clock is uniquely affected in frog or if the pitx3 perturbation inhibits the cellular changes that are necessary to rotation of presomitic cells. In Xenopus, pitx3 appears to inhibit the rotation of presomitic cell cohorts and to be necessary to the bilaterally symmetric expression of pitx2 in somites.
Collapse
Affiliation(s)
- Cristine Smoczer
- Biological Science, University of Windsor, 401 Sunset Avenue, Windsor, ON N9B 3P4, Canada
| | | | | | | | | | | |
Collapse
|
47
|
John LB, Trengove MC, Fraser FW, Yoong SH, Ward AC. Pegasus, the 'atypical' Ikaros family member, influences left-right asymmetry and regulates pitx2 expression. Dev Biol 2013; 377:46-54. [PMID: 23499657 DOI: 10.1016/j.ydbio.2013.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 02/21/2013] [Accepted: 02/21/2013] [Indexed: 12/12/2022]
Abstract
Members of the Ikaros family of zinc-finger transcription factors have been shown to be critical for immune and blood cell development. However, the role of the most divergent family member, Pegasus, has remained elusive, although it shows conservation to invertebrate Hunchback proteins that influence embryonic patterning through regulation of homeodomain genes. Zebrafish was employed as a relevant model to investigate the function of Pegasus since it possesses a single pegasus orthologue with high homology to its mammalian counterparts. During zebrafish embryogenesis pegasus transcripts were initially maternally-derived and later replaced by zygotic expression in the diencephalon, tectum, hindbrain, thymus, eye, and ultimately the exocrine pancreas and intestine. Morpholino-mediated knockdown of the zebrafish pegasus gene resulted in disrupted left-right asymmetry of the gut and pancreas. Molecular analysis indicated that zebrafish Pegasus localised to the nucleus in discrete non-nucleolar structures and bound the 'atypical' DNA sequence GN3GN2G, confirming its presumed role as a transcriptional regulator. In vivo transcriptome analysis identified candidate target genes, several of which encoded homeodomain transcription factors. One of these, pitx2, implicated in left-right asymmetry, possessed appropriate 'atypical' Pegasus binding sites in its promoter. Knockdown of Pegasus affected both the level and asymmetry of pitx2 expression, as well as disrupting the asymmetry of the lefty2 and spaw genes, explaining the perturbed left-right patterning in pegasus morphants. Collectively these results provide the first definitive insights into the in vivo role of Pegasus, supporting the notion that it acts as a broader regulator of development, with potential parallels to the related invertebrate Hunchback proteins.
Collapse
Affiliation(s)
- Liza B John
- School of Medicine and Molecular & Medical Research Strategic Research Centre, Deakin University, Waurn Ponds, Victoria, Australia
| | | | | | | | | |
Collapse
|
48
|
Basu M, Roy SS. Wnt/β-catenin pathway is regulated by PITX2 homeodomain protein and thus contributes to the proliferation of human ovarian adenocarcinoma cell, SKOV-3. J Biol Chem 2013; 288:4355-67. [PMID: 23250740 PMCID: PMC3567686 DOI: 10.1074/jbc.m112.409102] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 11/26/2012] [Indexed: 01/22/2023] Open
Abstract
Pituitary homeobox-2 (PITX2) plays a substantial role in the development of pituitary, heart, and brain. Although the role of PITX2 isoforms in embryonic development has been extensively studied, its possible involvement in regulating the Wnt signaling pathway has not been reported. Because the Wnt pathway is strongly involved in ovarian development and cancer, we focused on the possible association between PITX2 and Wnt pathway in ovarian carcinoma cells. Remarkably, we found that PITX2 interacts and regulates WNT2/5A/9A/6/2B genes of the canonical, noncanonical, or other pathways in the human ovarian cancer cell SKOV-3. Chromatin immunoprecipitation and promoter-reporter assays further indicated the significant association of PITX2 with WNT2 and WNT5A promoters. Detailed study further reveals that the PITX2 isoform specifically activates the canonical Wnt signaling pathway either directly or through Wnt ligands. Thus, the activated Wnt pathway subsequently enhances cell proliferation. Moreover, we found the activation of Wnt pathway reduces the expression of different FZD receptors that limit further Wnt activation, demonstrating the existence of an auto-regulatory feedback loop. In contrast, PITX2 could not activate the noncanonical pathway as the Wnt5A-specific ROR2 receptor does not express in SKOV-3 cells. Collectively, our findings demonstrated that, despite being a target of the canonical Wnt signaling pathway, PITX2 itself induces the same, thus leading to the activation of the cell cycle regulating genes as well as the proliferation of SKOV-3 cells. Collectively, we highlighted that the PITX2 and Wnt pathway exerts a positive feedback regulation, whereas frizzled receptors generate a negative feedback in this pathway. Our findings will help to understand the molecular mechanism of proliferation in ovarian cancer cells.
Collapse
Affiliation(s)
- Moitri Basu
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja Subodh Chandra Mullick Road, Kolkata 700032, India
| | - Sib Sankar Roy
- From the Cell Biology and Physiology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, 4 Raja Subodh Chandra Mullick Road, Kolkata 700032, India
| |
Collapse
|
49
|
Vandenberg LN, Lemire JM, Levin M. Serotonin has early, cilia-independent roles in Xenopus left-right patterning. Dis Model Mech 2013; 6:261-8. [PMID: 22899856 PMCID: PMC3529356 DOI: 10.1242/dmm.010256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 08/07/2012] [Indexed: 12/16/2022] Open
Abstract
Consistent left-right (LR) patterning of the heart and viscera is a crucial part of normal embryogenesis. Because errors of laterality form a common class of birth defects, it is important to understand the molecular mechanisms and stage at which LR asymmetry is initiated. Frog embryos are a system uniquely suited to analysis of the mechanisms involved in orientation of the LR axis because of the many genetic and pharmacological tools available for use and the fate-map and accessibility of early blastomeres. Two major models exist for the origin of LR asymmetry and both implicate pre-nervous serotonergic signaling. In the first, the charged serotonin molecule is instructive for LR patterning; it is redistributed asymmetrically along the LR axis and signals intracellularly on the right side at cleavage stages. A second model suggests that serotonin is a permissive factor required to specify the dorsal region of the embryo containing chiral cilia that generate asymmetric fluid flow during neurulation, a much later process. We performed theory-neutral experiments designed to distinguish between these models. The results uniformly support a role for serotonin in the cleavage-stage embryo, long before the appearance of cilia, in ventral right blastomeres that do not contribute to the ciliated organ.
Collapse
Affiliation(s)
- Laura N. Vandenberg
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155, USA
| | - Joan M. Lemire
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155, USA
| | - Michael Levin
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155, USA
| |
Collapse
|
50
|
Waite MR, Skidmore JM, Micucci JA, Shiratori H, Hamada H, Martin JF, Martin DM. Pleiotropic and isoform-specific functions for Pitx2 in superior colliculus and hypothalamic neuronal development. Mol Cell Neurosci 2013; 52:128-39. [PMID: 23147109 PMCID: PMC3540135 DOI: 10.1016/j.mcn.2012.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 10/01/2012] [Accepted: 11/02/2012] [Indexed: 02/01/2023] Open
Abstract
Transcriptional regulation of gene expression during development is critical for proper neuronal differentiation and migration. Alternative splicing and differential isoform expression have been demonstrated for most mammalian genes, but their specific contributions to gene function are not well understood. In mice, the transcription factor gene Pitx2 is expressed as three different isoforms (PITX2A, PITX2B, and PITX2C) which have unique amino termini and common DNA binding homeodomains and carboxyl termini. The specific roles of these isoforms in neuronal development are not known. Here we report the onset of Pitx2ab and Pitx2c isoform-specific expression by E9.5 in the developing mouse brain. Using isoform-specific Pitx2 deletion mouse strains, we show that collicular neuron migration requires PITX2AB and that collicular GABAergic differentiation and targeting of hypothalamic projections require unique Pitx2 isoform dosage. These results provide insights into Pitx2 dosage and isoform-specific requirements underlying midbrain and hypothalamic development.
Collapse
Affiliation(s)
- Mindy R Waite
- Cellular and Molecular Biology Graduate Program, 2966 Taubman Medical Library, University of Michigan, Ann Arbor, MI 48109-0619, USA.
| | | | | | | | | | | | | |
Collapse
|