1
|
VanDerMolen KR, Newman MA, Breen PC, Gao Y, Huff LA, Dowen RH. Non-cell-autonomous regulation of mTORC2 by Hedgehog signaling maintains lipid homeostasis. Cell Rep 2025; 44:115191. [PMID: 39786994 PMCID: PMC11834565 DOI: 10.1016/j.celrep.2024.115191] [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: 06/24/2024] [Revised: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Organisms allocate energetic resources between essential cellular processes to maintain homeostasis and, in turn, maximize fitness. The nutritional regulators of energy homeostasis have been studied in detail; however, how developmental signals might impinge on these pathways to govern metabolism is poorly understood. Here, we identify a non-canonical role for Hedgehog (Hh), a classic regulator of development, in maintaining intestinal lipid homeostasis in Caenorhabditis elegans. We demonstrate, using C. elegans and mouse hepatocytes, that Hh metabolic regulation does not occur through the canonical Hh transcription factor TRA-1/GLI, but rather via non-canonical signaling that engages mammalian target of rapamycin complex 2 (mTORC2). Hh mutants display impaired lipid homeostasis, decreased growth, and upregulation of autophagy factors, mimicking loss of mTORC2. Additionally, we find that Hh inhibits p38 MAPK signaling in parallel to mTORC2 activation to modulate lipid homeostasis. Our findings reveal a non-canonical role for Hh signaling in lipid metabolism via regulation of core homeostatic pathways.
Collapse
Affiliation(s)
- Kylie R VanDerMolen
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin A Newman
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter C Breen
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yunjing Gao
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Laura A Huff
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robert H Dowen
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
2
|
Bhaskar M, Satheesan A, Basu A. Low-density Lipoprotein Receptor is an important host factor in flaviviral entry and replication in neurons. Biochem Biophys Res Commun 2025; 743:151160. [PMID: 39689643 DOI: 10.1016/j.bbrc.2024.151160] [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: 09/16/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Flaviviruses, which are transmitted by mosquitoes, are arthropod-borne infections that are pathogenic to both humans and animals, posing a significant global threat to public health. So far, various endocytic pathways have been reported for flaviviral entry; however, the role of cellular factors in viral replication and entry remains uncertain. Here in this study, we identified the role of Low-density lipoprotein receptor, which has long been established as a cholesterol carrier for neurons but remained unexplored as an essential host factor for JEV/WNV replication. To explore this, we utilized 10-day old BALB/c pups and two neuronal cell lines, NSC34 and HT22, both of different origin, as experimental models. Transient knockdown of LDLR gene in vitro using siRNA-mediated gene silencing drastically reduced viral specific transcripts and proteins upon viral incubation. Moreover, flaviviral binding and internalization were significantly compromised upon infection in LDLR-transfected cells when compared with non-specific eGFP-transfected cells. Antibody neutralization experiments using LDLR-specific polyclonal antibody significantly reduced viral entry in vitro, suggesting the role of LDLR as an important cell attachment factor for JEV and WNV uptake. Furthermore, ectopic expression of LDLR via plasmid transfection led to significant increase in virus replication in cells, indicating significant role of LDLR in flavivirus replication beside acting as an active attachment factor for JEV and WNV. Overall, our results indicate that LDLR act as novel host factor involved in both flaviviral entry and replication, thus serving as a suitable candidate for antiviral research.
Collapse
Affiliation(s)
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| |
Collapse
|
3
|
Liang Y, Zhong G, Li Y, Ren M, Wang A, Ying M, Liu C, Guo Y, Zhang D. Comprehensive Analysis and Experimental Validation of the Parkinson's Disease Lysosomal Gene ACP2 and Pan-cancer. Biochem Genet 2024; 62:4408-4431. [PMID: 38310198 DOI: 10.1007/s10528-023-10652-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/27/2023] [Indexed: 02/05/2024]
Abstract
The pivotal role of lysosomal function in preserving neuronal homeostasis is recognized, with its dysfunction being implicated in neurodegenerative processes, notably in Parkinson's disease (PD). Yet, the molecular underpinnings of lysosome-related genes (LRGs) in the context of PD remain partially elucidated. We collected RNA-seq data from the brain substantia nigra of 30 PD patients and 20 normal subjects from the GEO database. We obtained molecular classification clusters from the screened lysosomal expression patterns. The lysosome-related diagnostic model of Parkinson's disease was constructed by XGBoost and Random Forest. And we validated the expression patterns of signature LRGs in the diagnostic model by constructing a PD rat model. Finally, the linkage between PD and cancer through signature genes was explored. The expression patterns of the 33 LRGs screened can be divided into two groups of PD samples, enabling exploration of the variance in biological processes and immune elements. Cluster A had a higher disease severity. Subsequently, critical genes were sieved through the application of machine learning methodologies culminating in the identification of two intersecting feature genes (ACP2 and LRP2). A PD risk prediction model was constructed grounded on these signature genes. The model's validity was assessed through nomogram evaluation, which demonstrated robust confidence validity. Then we analyzed the correlation analysis, immune in-filtration, biological function, and rat expression validation of the two genes with common pathogenic genes in Parkinson's disease, indicating that these two genes play an important role in the pathogenesis of PD. We then selected ACP2, which had a significant immune infiltration correlation, as the entry gene for the pan-cancer analysis. The pan-cancer analysis revealed that ACP2 has profound associations with prognostic indicators, immune infiltration, and tumor-related regulatory processes across various neoplasms, suggesting its potential as a therapeutic target in a range of human diseases, including PD and cancers. Our study comprehensively analyzed the molecular grouping of LRGs expression patterns in Parkinson's disease, and the disease progression was more severe in cluster A. And the PD diagnosis model related to LRGs is constructed. Finally, ACP2 is a potential target for the relationship between Parkinson's disease and tumor.
Collapse
Affiliation(s)
- Yu Liang
- School of Clinical Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Guangshang Zhong
- School of Clinical Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Yangyang Li
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Mingxin Ren
- School of Clinical Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Ao Wang
- School of Clinical Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233000, China
| | - Mengjiao Ying
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Changqing Liu
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| | - Yu Guo
- School of Clinical Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233000, China.
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| | - Ding Zhang
- School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
4
|
VanDerMolen KR, Newman MA, Breen PC, Huff LA, Dowen RH. Non-cell-autonomous regulation of mTORC2 by Hedgehog signaling maintains lipid homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592795. [PMID: 38766075 PMCID: PMC11100691 DOI: 10.1101/2024.05.06.592795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Organisms must appropriately allocate energetic resources between essential cellular processes to maintain homeostasis and in turn, maximize fitness. The nutritional and homeostatic regulators of energy homeostasis have been studied in detail; however, how developmental signals might impinge on these pathways to govern cellular metabolism is poorly understood. Here, we identify a non-canonical role for Hedgehog (Hh), a classic regulator of development, in maintaining intestinal lipid homeostasis in C. elegans . We find that expression of two Hh ligands, GRD-3 and GRD-4, is controlled by the LIN-29/EGR transcription factor in the hypodermis, where the Hh secretion factor CHE-14/Dispatched also facilitates non-cell autonomous Hh signaling. We demonstrate, using C. elegans and mouse hepatocytes, that Hh metabolic regulation does not occur through the canonical Hh transcription factor, TRA-1/GLI, but rather through non-canonical signaling that engages mTOR Complex 2 (mTORC2) in the intestine. Hh mutants display impaired lipid homeostasis, including reduced lipoprotein synthesis and fat accumulation, decreased growth, and upregulation of autophagy factors, mimicking loss of mTORC2. Additionally, we found that Hh inhibits p38 MAPK signaling in parallel to mTORC2 activation and that both pathways act together to modulate of lipid homeostasis. Our findings show a non-canonical role for Hedgehog signaling in lipid metabolism via regulation of core homeostatic pathways and reveal a new mechanism by which developmental timing events govern metabolic decisions.
Collapse
|
5
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [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: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
6
|
da Cunha JI, Barauna AMD, Garcez RC. Prechordal structures act cooperatively in early trabeculae development of gnathostome skull. Cells Dev 2023; 176:203879. [PMID: 37844659 DOI: 10.1016/j.cdev.2023.203879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
The vertebrate skull is formed by mesoderm and neural crest (NC) cells. The mesoderm contributes to the skull chordal domain, with the notochord playing an essential role in this process. The NC contributes to the skull prechordal domain, prompting investigation into the embryonic structures involved in prechordal neurocranium cartilage formation. The trabeculae cartilage, a structure of the prechordal neurocranium, arises at the convergence of prechordal plate (PCP), ventral midline (VM) cells of the diencephalon, and dorsal oral ectoderm. This study examines the molecular participation of these embryonic structures in gnathostome trabeculae development. PCP-secreted SHH induces its expression in VM cells of the diencephalon, initiating a positive feedback loop involving SIX3 and GLI1. SHH secreted by the VM cells of the diencephalon acts on the dorsal oral ectoderm, stimulating condensation of NC cells to form trabeculae. SHH from the prechordal region affects the expression of SOX9 in NC cells. BMP7 and SHH secreted by PCP induce NKX2.1 expression in VM cells of the diencephalon, but this does not impact trabeculae formation. Molecular cooperation between PCP, VM cells of the diencephalon, and dorsal oral ectoderm is crucial for craniofacial development by NC cells in the prechordal domain.
Collapse
Affiliation(s)
- Jaqueline Isoppo da Cunha
- Graduate Program of Cell and Developmental Biology, Federal University of Santa Catarina, Florianopolis, SC, Brazil; Stem Cell and Tissue Regeneration Laboratory (LACERT), Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Alessandra Maria Duarte Barauna
- Graduate Program of Cell and Developmental Biology, Federal University of Santa Catarina, Florianopolis, SC, Brazil; Stem Cell and Tissue Regeneration Laboratory (LACERT), Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Ricardo Castilho Garcez
- Graduate Program of Cell and Developmental Biology, Federal University of Santa Catarina, Florianopolis, SC, Brazil; Stem Cell and Tissue Regeneration Laboratory (LACERT), Federal University of Santa Catarina, Florianopolis, SC, Brazil; Department of Cell Biology, Embryology, and Genetics, Federal University of Santa Catarina, Florianopolis, SC, Brazil.
| |
Collapse
|
7
|
Keuls RA, Finnell RH, Parchem RJ. Maternal metabolism influences neural tube closure. Trends Endocrinol Metab 2023; 34:539-553. [PMID: 37468429 PMCID: PMC10529122 DOI: 10.1016/j.tem.2023.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023]
Abstract
Changes in maternal nutrient availability due to diet or disease significantly increase the risk of neural tube defects (NTDs). Because the incidence of metabolic disease continues to rise, it is urgent that we better understand how altered maternal nutrient levels can influence embryonic neural tube development. Furthermore, primary neurulation occurs before placental function during a period of histiotrophic nutrient exchange. In this review we detail how maternal metabolites are transported by the yolk sac to the developing embryo. We discuss recent advances in understanding how altered maternal levels of essential nutrients disrupt development of the neuroepithelium, and identify points of intersection between metabolic pathways that are crucial for NTD prevention.
Collapse
Affiliation(s)
- Rachel A Keuls
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine. Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard H Finnell
- Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Precision Environmental Health, Department of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ronald J Parchem
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine. Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Beenken A, Cerutti G, Brasch J, Guo Y, Sheng Z, Erdjument-Bromage H, Aziz Z, Robbins-Juarez SY, Chavez EY, Ahlsen G, Katsamba PS, Neubert TA, Fitzpatrick AWP, Barasch J, Shapiro L. Structures of LRP2 reveal a molecular machine for endocytosis. Cell 2023; 186:821-836.e13. [PMID: 36750096 PMCID: PMC9993842 DOI: 10.1016/j.cell.2023.01.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023]
Abstract
The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2 or megalin) is representative of the phylogenetically conserved subfamily of giant LDL receptor-related proteins, which function in endocytosis and are implicated in diseases of the kidney and brain. Here, we report high-resolution cryoelectron microscopy structures of LRP2 isolated from mouse kidney, at extracellular and endosomal pH. The structures reveal LRP2 to be a molecular machine that adopts a conformation for ligand binding at the cell surface and for ligand shedding in the endosome. LRP2 forms a homodimer, the conformational transformation of which is governed by pH-sensitive sites at both homodimer and intra-protomer interfaces. A subset of LRP2 deleterious missense variants in humans appears to impair homodimer assembly. These observations lay the foundation for further understanding the function and mechanism of LDL receptors and implicate homodimerization as a conserved feature of the LRP receptor subfamily.
Collapse
Affiliation(s)
- Andrew Beenken
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Gabriele Cerutti
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Julia Brasch
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Zainab Aziz
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Estefania Y Chavez
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Goran Ahlsen
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Phinikoula S Katsamba
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony W P Fitzpatrick
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Columbia University George M. O'Brien Urology Center, New York, NY 10032, USA.
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
9
|
LRP2 contributes to planar cell polarity-dependent coordination of motile cilia function. Cell Tissue Res 2023; 392:535-551. [PMID: 36764939 PMCID: PMC10172251 DOI: 10.1007/s00441-023-03757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 02/12/2023]
Abstract
Motile cilia are protruding organelles on specialized epithelia that beat in a synchronous fashion to propel extracellular fluids. Coordination and orientation of cilia beating on individual cells and across tissues is a complex process dependent on planar cell polarity (PCP) signaling. Asymmetric sorting of PCP pathway components, essential to establish planar polarity, involves trafficking along the endocytic path, but the underlying regulatory processes remain incompletely understood. Here, we identified the endocytic receptor LRP2 as regulator of PCP component trafficking in ependyma, a multi-ciliated cell type that is involved in facilitating flow of the cerebrospinal fluid in the brain ventricular system. Lack of receptor expression in gene-targeted mice results in a failure to sort PCP core proteins to the anterior or posterior cell side and, consequently, in the inability to coordinate cilia arrangement and to aligned beating (loss of rotational and translational polarity). LRP2 deficiency coincides with a failure to sort NHERF1, a cytoplasmic LRP2 adaptor to the anterior cell side. As NHERF1 is essential to translocate PCP core protein Vangl2 to the plasma membrane, these data suggest a molecular mechanism whereby LRP2 interacts with PCP components through NHERF1 to control their asymmetric sorting along the endocytic path. Taken together, our findings identified the endocytic receptor LRP2 as a novel regulator of endosomal trafficking of PCP proteins, ensuring their asymmetric partition and establishment of translational and rotational planar cell polarity in the ependyma.
Collapse
|
10
|
Tournier B, Aucagne R, Truntzer C, Fournier C, Ghiringhelli F, Chapusot C, Martin L, Bouvier AM, Manfredi S, Jooste V, Callanan MB, Lepage C. Integrative Clinical and DNA Methylation Analyses in a Population-Based Cohort Identifies CDH17 and LRP2 as Risk Recurrence Factors in Stage II Colon Cancer. Cancers (Basel) 2022; 15:cancers15010158. [PMID: 36612154 PMCID: PMC9817957 DOI: 10.3390/cancers15010158] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Stage II colon cancer (CC), although diagnosed early, accounts for 16% of CC deaths. Predictors of recurrence risk could mitigate this but are currently lacking. By using a DNA methylation-based clinical screening in real-world (n = 383) and in TCGA-derived cohorts of stage II CC (n = 134), we have devised a novel 40 CpG site-based classifier that can segregate stage II CC into four previously undescribed disease sub-classes that are characterised by distinct molecular features, including activation of MYC/E2F-dependant proliferation signatures. By multivariate analyses, hypermethylation of 2 CpG sites at genes CDH17 and LRP2, respectively, was found to independently confer either significantly increased (CDH17; p-value, 0.0203) or reduced (LRP2; p-value, 0.0047) risk of CC recurrence. Functional enrichment and immune cell infiltration analyses, on RNAseq data from the TCGA cohort, revealed cases with hypermethylation at CDH17 to be enriched for KRAS, epithelial-mesenchymal transition and inflammatory functions (via IL2/STAT5), associated with infiltration by 'exhausted' T cells. By contrast, LRP2 hypermethylated cases showed enrichment for mTORC1, DNA repair pathways and activated B cell signatures. These findings will be of value for improving personalised care paths and treatment in stage II CC patients.
Collapse
Affiliation(s)
- Benjamin Tournier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Romain Aucagne
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
| | - Caroline Truntzer
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Centre Georges-François Leclerc (CGFL), 21000 Dijon, France
| | - Cyril Fournier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
| | - François Ghiringhelli
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
- Centre Georges-François Leclerc (CGFL), 21000 Dijon, France
| | - Caroline Chapusot
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Laurent Martin
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Pathology, Dijon University Hospital, 21000 Dijon, France
| | - Anne Marie Bouvier
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
| | - Sylvain Manfredi
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
| | - Valérie Jooste
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
| | - Mary B. Callanan
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO) and CRIGEN (Crispr Functional Genomics), Dijon University Hospital, 21000 Dijon, France
- Genetics and Immunology Medical Institute (GIMI), 21000 Dijon, France
- Correspondence: (M.B.C.); (C.L.)
| | - Côme Lepage
- Faculty of Health Sciences, University of Burgundy, 21000 Dijon, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1231, 21000 Dijon, France
- Department of Hepato-Gastroenterology and Digestive Oncology, Dijon University Hospital, 21000 Dijon, France
- Correspondence: (M.B.C.); (C.L.)
| |
Collapse
|
11
|
Williams DM, Gungordu L, Jackson-Crawford A, Lowe M. Assessment of endocytic traffic and Ocrl function in the developing zebrafish neuroepithelium. J Cell Sci 2022; 135:276669. [PMID: 35979861 PMCID: PMC9592051 DOI: 10.1242/jcs.260339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/11/2022] [Indexed: 12/05/2022] Open
Abstract
Endocytosis allows cells to internalise a wide range of molecules from their environment and to maintain their plasma membrane composition. It is vital during development and for maintenance of tissue homeostasis. The ability to visualise endocytosis in vivo requires suitable assays to monitor the process. Here, we describe imaging-based assays to visualise endocytosis in the neuroepithelium of living zebrafish embryos. Injection of fluorescent tracers into the brain ventricles followed by live imaging was used to study fluid-phase or receptor-mediated endocytosis, for which we used receptor-associated protein (RAP, encoded by Lrpap1) as a ligand for low-density lipoprotein receptor-related protein (LRP) receptors. Using dual-colour imaging combined with expression of endocytic markers, it is possible to track the progression of endocytosed tracers and to monitor trafficking dynamics. Using these assays, we reveal a role for the Lowe syndrome protein Ocrl in endocytic trafficking within the neuroepithelium. We also found that the RAP-binding receptor Lrp2 (encoded by lrp2a) appears to contribute only partially to neuroepithelial RAP endocytosis. Altogether, our results provide a basis to track endocytosis within the neuroepithelium in vivo and support a role for Ocrl in this process. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Daniel M Williams
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Lale Gungordu
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Anthony Jackson-Crawford
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
12
|
Mai S, Zhu X, Wan EYC, Wu S, Yonathan JN, Wang J, Li Y, Ma JYW, Zuo B, Tse DYY, Lo PC, Wang X, Chan KM, Wu DM, Xiong W. Postnatal eye size in mice is controlled by SREBP2-mediated transcriptional repression of Lrp2 and Bmp2. Development 2022; 149:276005. [PMID: 35833708 PMCID: PMC9382895 DOI: 10.1242/dev.200633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/20/2022] [Indexed: 11/20/2022]
Abstract
Eye size is a key parameter of visual function, but the precise mechanisms of eye size control remain poorly understood. Here, we discovered that the lipogenic transcription factor sterol regulatory element-binding protein 2 (SREBP2) has an unanticipated function in the retinal pigment epithelium (RPE) to promote eye size in postnatal mice. SREBP2 transcriptionally represses low density lipoprotein receptor-related protein 2 (Lrp2), which has been shown to restrict eye overgrowth. Bone morphogenetic protein 2 (BMP2) is the downstream effector of Srebp2 and Lrp2, and Bmp2 is suppressed by SREBP2 transcriptionally but activated by Lrp2. During postnatal development, SREBP2 protein expression in the RPE decreases whereas that of Lrp2 and Bmp2 increases as the eye growth rate reduces. Bmp2 is the key determinant of eye size such that its level in mouse RPE inversely correlates with eye size. Notably, RPE-specific Bmp2 overexpression by adeno-associated virus effectively prevents the phenotypes caused by Lrp2 knock out. Together, our study shows that rapid postnatal eye size increase is governed by an RPE-derived signaling pathway, which consists of both positive and negative regulators of eye growth.
Collapse
Affiliation(s)
- Shuyi Mai
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China.,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Xiaoxuan Zhu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Esther Yi Ching Wan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Shengyu Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | | | - Jun Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Li
- College of Information and Computer, Taiyuan University of Technology, 030024 Taiyuan, China
| | - Jessica Yuen Wuen Ma
- Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
| | - Bing Zuo
- Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
| | - Dennis Yan-Yin Tse
- Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China.,Research Centre for SHARP Vision, Hong Kong Polytechnic University, Hong Kong, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| | - David M Wu
- Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Wenjun Xiong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| |
Collapse
|
13
|
Li C, Ding Y, Zhang X, Hua K. Integrated in silico analysis of LRP2 mutations to immunotherapy efficacy in pan-cancer cohort. Discov Oncol 2022; 13:65. [PMID: 35834061 PMCID: PMC9283634 DOI: 10.1007/s12672-022-00528-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Immunotherapy has emerged as a novel therapy, while many patients are refractory. Although, several biomarkers have been identified as predictive biomarkers for immunotherapy, such as tumor specific genes, PD-1/PD-L1, tumor mutation burn (TMB), and microsatellite instability (MSI), results remain unsatisfactory. The aim of this study is to evaluate the value of LRP2 mutations in predicating cancer immunotherapy. METHODS We investigated the characteristics of low-density lipoprotein receptor-related protein 2 (LRP2) mutation in the cancer genome atlas (TCGA) and explored the potential association of LRP2 mutations with immunotherapy. Characteristics of LRP2 mutations in 33 cancer types were analyzed using large-scale public data. The association of LRP2 mutations with immune cell infiltration and immunotherapy efficacy was evaluated. Finally, a LPR2 mutation signature (LMS) was developed and validated by TCGA-UCEC and pan-cancer cohorts. Furthermore, we demonstrated the predictive power of LMS score in independent immunotherapy cohorts by performing a meta-analysis. RESULTS Our results revealed that patients with LRP2 mutant had higher TMB and MSI compared with patients without LRP2 mutations. LRP2 mutations were associated with high levels of immune cells infiltration, immune-related genes expression and enrichment of immune related signaling pathways. Importantly, LRP2-mutated patients had a long overall survival (OS) after immunotherapy. In the endometrial cancer (EC) cohort, we found that patients with LRP2 mutations belonged to the POLE and MSI-H type and had a better prognosis. Finally, we developed a LRP2 mutations signature (LMS), that was significantly associated with prognosis in patients receiving immunotherapy. CONCLUSION These results indicated that LRP2 mutations can serve as a biomarker for personalized tumor immunotherapy. Importantly, LMS is a potential predictor of patients' prognosis after immunotherapy.
Collapse
Affiliation(s)
- Chunbo Li
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 FangXie Road, Shanghai, 200011, China
| | - Yan Ding
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 FangXie Road, Shanghai, 200011, China
| | - Xuyin Zhang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 FangXie Road, Shanghai, 200011, China.
| | - Keqin Hua
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 419 FangXie Road, Shanghai, 200011, China.
| |
Collapse
|
14
|
Nguyen TD, Truong ME, Reiter JF. The Intimate Connection Between Lipids and Hedgehog Signaling. Front Cell Dev Biol 2022; 10:876815. [PMID: 35757007 PMCID: PMC9222137 DOI: 10.3389/fcell.2022.876815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/13/2022] [Indexed: 01/19/2023] Open
Abstract
Hedgehog (HH) signaling is an intercellular communication pathway involved in directing the development and homeostasis of metazoans. HH signaling depends on lipids that covalently modify HH proteins and participate in signal transduction downstream. In many animals, the HH pathway requires the primary cilium, an organelle with a specialized protein and lipid composition. Here, we review the intimate connection between HH signaling and lipids. We highlight how lipids in the primary cilium can create a specialized microenvironment to facilitate signaling, and how HH and components of the HH signal transduction pathway use lipids to communicate between cells.
Collapse
Affiliation(s)
- Thi D. Nguyen
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Melissa E. Truong
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
15
|
Luo S, Xu H, Yang L, Gong X, Shen J, Chen X, Wu Z. Quantitative proteomics analysis of human vitreous in rhegmatogenous retinal detachment associated with choroidal detachment by data-independent acquisition mass spectrometry. Mol Cell Biochem 2022; 477:1849-1863. [PMID: 35332395 DOI: 10.1007/s11010-022-04409-0] [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: 08/04/2021] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The prognosis of rhegmatogenous retinal detachment (RRD) with choroidal detachment (RRDCD) is often poor and complicated. This study focused on the identification of the characteristic proteins and signal pathways associated with the etiology of RRDCD and to provide guidance for diagnosis and treatment of RRDCD. In this study, vitreous humor samples were obtained from 16 RRDCD patients, 14 with RRD, 12 with idiopathic epiretinal macular membrane (IEMM), and 5 healthy controls from donated corpse eyes. Data-independent acquisition mass spectrometry and bioinformatics analysis were employed to identify differentially expressed proteins (DEPs). In the vitreous humor, 14,842 peptides were identified. Patients with RRDCD had 249 DEPs (93 upregulated and 156 downregulated), with 89 in patients with RRD and 61 in patients with IEMM. Enrichment analysis of the GO and Kyoto Encyclopedia of Genes and Genomes DEP databases indicated functional clusters related to inflammation and immunity, protein degradation and absorption, cell adhesion molecules (CAMs), the hedgehog signaling pathway, and lipid metabolism. Weighted gene co-expression network analysis showed that DEPs with positive co-expression of RRDCD participated in immune-related pathways led by the complement and coagulation cascade, whereas DEPs with negative co-expression of RRDCD participated in protein degradation and absorption, CAMs, and the hedgehog signaling pathway. In summary, our study provides important clues and the theoretical basis for exploring the pathogenesis, progression, and prognosis of ocular fundus disease.
Collapse
Affiliation(s)
- Shasha Luo
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China.,Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, People's Republic of China
| | - Huiyan Xu
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China.,Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, People's Republic of China
| | - Lufei Yang
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Xuechun Gong
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Jinyan Shen
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Xuan Chen
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China
| | - Zhifeng Wu
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, 68 Zhongshan Road, Wuxi, 214002, Jiangsu Province, People's Republic of China. .,Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, People's Republic of China.
| |
Collapse
|
16
|
Barratt KS, Drover KA, Thomas ZM, Arkell RM. Patterning of the antero-ventral mammalian brain: Lessons from holoprosencephaly comparative biology in man and mouse. WIREs Mech Dis 2022; 14:e1552. [PMID: 35137563 DOI: 10.1002/wsbm.1552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Adult form and function are dependent upon the activity of specialized signaling centers that act early in development at the embryonic midline. These centers instruct the surrounding cells to adopt a positional fate and to form the patterned structures of the phylotypic embryo. Abnormalities in these processes have devastating consequences for the individual, as exemplified by holoprosencephaly in which anterior midline development fails, leading to structural defects of the brain and/or face. In the 25 years since the first association between human holoprosencephaly and the sonic hedgehog gene, a combination of human and animal genetic studies have enhanced our understanding of the genetic and embryonic causation of this congenital defect. Comparative biology has extended the holoprosencephaly network via the inclusion of gene mutations from multiple signaling pathways known to be required for anterior midline formation. It has also clarified aspects of holoprosencephaly causation, showing that it arises when a deleterious variant is present within a permissive genome, and that environmental factors, as well as embryonic stochasticity, influence the phenotypic outcome of the variant. More than two decades of research can now be distilled into a framework of embryonic and genetic causation. This framework means we are poised to move beyond our current understanding of variants in signaling pathway molecules. The challenges now at the forefront of holoprosencephaly research include deciphering how the mutation of genes involved in basic cell processes can also cause holoprosencephaly, determining the important constituents of the holoprosencephaly permissive genome, and identifying environmental compounds that promote holoprosencephaly. This article is categorized under: Congenital Diseases > Stem Cells and Development Congenital Diseases > Genetics/Genomics/Epigenetics Congenital Diseases > Molecular and Cellular Physiology Congenital Diseases > Environmental Factors.
Collapse
Affiliation(s)
- Kristen S Barratt
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kyle A Drover
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Zoe M Thomas
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Ruth M Arkell
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
17
|
Wang Y, Chen X, Jiang T, Gu Y, Zhang X, Yuan W, Zhao A, Li R, Wang Z, Hu Z, Liu H. Expanding the phenotypic spectrum of mutations in LRP2: a novel candidate gene of non-syndromic familial comitant strabismus. J Transl Med 2021; 19:495. [PMID: 34872573 PMCID: PMC8647414 DOI: 10.1186/s12967-021-03155-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
Background Comitant strabismus (CS) is a heterogeneous disorder that is a major contributing factor to unilateral childhood-onset visual impairment. Studies have confirmed that genetic factors play an important role in the development of CS. The aim of this study was to identify the genetic cause of non-syndromic familial CS. Methods Fourteen unrelated CS families were recruited for the study. Twelve affected and 2 unaffected individuals from a large four-generation family (CS08) were selected to perform whole genome-wide linkage analysis. Parallel whole-exome sequencing (WES) was conducted in the same family (9 patients and 1 unaffected member) and 31 additional CS cases from 13 other unrelated families. Sanger sequencing was used to determine whether any of the remaining variants co-segregated with the disease phenotype in the corresponding family. Results Based on linkage analysis, CS in family CS08 mapped to a novel region of 34.17 centimorgan (cM) on chromosome 2q22.3-2q32.1 between markers D2S151 and D2S364, with a maximum log odds (LOD) score of 3.54 (theta = 0) at D2S142. Parallel WES identified a heterozygous variant, LRP2 c.335 A > G (p.Q112R), located in such a linkage interval that completely co-segregated with the disease in the family. Furthermore, another novel heterozygous variant (c.7274A > G, p.D2425G) in LRP2 that co-segregated was detected in 2 additional affected individuals from another unrelated family by WES. Both variants are predicted to be damaging by PolyPhen-2, SIFT and MutationTaster, and were absent in 100 ethnically matched normal controls. Conclusion LRP2 is a novel candidate genetic cause of non-syndromic familial CS. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03155-z.
Collapse
Affiliation(s)
- Yue Wang
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.,Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Xuejuan Chen
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.,Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Tao Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Yayun Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China
| | - Xiaohan Zhang
- Department of Ophthalmology, Wuxi Children's Hospital, Wuxi, China
| | - Wenwen Yuan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Andi Zhao
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Rui Li
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Zijin Wang
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Rd, NanjingNanjing, 211166, China.
| | - Hu Liu
- Department of Ophthalmology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China.
| |
Collapse
|
18
|
Marczenke M, Sunaga-Franze DY, Popp O, Althaus IW, Sauer S, Mertins P, Christ A, Allen BL, Willnow TE. GAS1 is required for NOTCH-dependent facilitation of SHH signaling in the ventral forebrain neuroepithelium. Development 2021; 148:272617. [PMID: 34698766 PMCID: PMC8627604 DOI: 10.1242/dev.200080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
Growth arrest-specific 1 (GAS1) acts as a co-receptor to patched 1, promoting sonic hedgehog (SHH) signaling in the developing nervous system. GAS1 mutations in humans and animal models result in forebrain and craniofacial malformations, defects ascribed to a function for GAS1 in SHH signaling during early neurulation. Here, we confirm loss of SHH activity in the forebrain neuroepithelium in GAS1-deficient mice and in induced pluripotent stem cell-derived cell models of human neuroepithelial differentiation. However, our studies document that this defect can be attributed, at least in part, to a novel role for GAS1 in facilitating NOTCH signaling, which is essential to sustain a persistent SHH activity domain in the forebrain neuroepithelium. GAS1 directly binds NOTCH1, enhancing ligand-induced processing of the NOTCH1 intracellular domain, which drives NOTCH pathway activity in the developing forebrain. Our findings identify a unique role for GAS1 in integrating NOTCH and SHH signal reception in neuroepithelial cells, and they suggest that loss of GAS1-dependent NOTCH1 activation contributes to forebrain malformations in individuals carrying GAS1 mutations.
Collapse
Affiliation(s)
- Maike Marczenke
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Department of Biology, Chemistry and Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany
| | | | - Oliver Popp
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Irene W Althaus
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sascha Sauer
- Genomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Annabel Christ
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Thomas E Willnow
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
19
|
Abrams SR, Reiter JF. Ciliary Hedgehog signaling regulates cell survival to build the facial midline. eLife 2021; 10:e68558. [PMID: 34672258 PMCID: PMC8592574 DOI: 10.7554/elife.68558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Craniofacial defects are among the most common phenotypes caused by ciliopathies, yet the developmental and molecular etiology of these defects is poorly understood. We investigated multiple mouse models of human ciliopathies (including Tctn2, Cc2d2a, and Tmem231 mutants) and discovered that each displays hypotelorism, a narrowing of the midface. As early in development as the end of gastrulation, Tctn2 mutants displayed reduced activation of the Hedgehog (HH) pathway in the prechordal plate, the head organizer. This prechordal plate defect preceded a reduction of HH pathway activation and Shh expression in the adjacent neurectoderm. Concomitant with the reduction of HH pathway activity, Tctn2 mutants exhibited increased cell death in the neurectoderm and facial ectoderm, culminating in a collapse of the facial midline. Enhancing HH signaling by decreasing the gene dosage of a negative regulator of the pathway, Ptch1, decreased cell death and rescued the midface defect in both Tctn2 and Cc2d2a mutants. These results reveal that ciliary HH signaling mediates communication between the prechordal plate and the neurectoderm to provide cellular survival cues essential for development of the facial midline.
Collapse
Affiliation(s)
- Shaun R Abrams
- Department of Biochemistry and Biophysics, Cardiovascular Research InstituteSan FranciscoUnited States
- Oral and Craniofacial Sciences Program, School of DentistrySan FranciscoUnited States
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research InstituteSan FranciscoUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| |
Collapse
|
20
|
Pillai VV, Koganti PP, Kei TG, Gurung S, Butler WR, Selvaraj V. Efficient induction and sustenance of pluripotent stem cells from bovine somatic cells. Biol Open 2021; 10:272681. [PMID: 34719702 PMCID: PMC8565620 DOI: 10.1242/bio.058756] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Although derivation of naïve bovine embryonic stem cells is unachieved, the possibility for generation of bovine induced pluripotent stem cells (biPSCs) has been generally reported. However, attempts to sustain biPSCs by promoting self-renewal have not been successful. Methods established for maintaining murine and human induced pluripotent stem cells (iPSCs) do not support self-renewal of iPSCs for any bovid species. In this study, we examined methods to enhance complete reprogramming and concurrently investigated signaling relevant to pluripotency of the bovine blastocyst inner cell mass (ICM). First, we identified that forced expression of SV40 large T antigen together with the reprogramming genes (OCT4, SOX2, KLF4 and MYC) substantially enhanced the reprogramming efficacy of bovine fibroblasts to biPSCs. Second, we uncovered that TGFβ signaling is actively perturbed in the ICM. Inhibition of ALK4/5/7 to block TGFβ/activin/nodal signaling together with GSK3β and MEK1/2 supported robust in vitro self-renewal of naïve biPSCs with unvarying colony morphology, steady expansion, expected pluripotency gene expression and committed differentiation plasticity. Core similarities between biPSCs and stem cells of the 16-cell-stage bovine embryo indicated a stable ground state of pluripotency; this allowed us to reliably gain predictive understanding of signaling in bovine pluripotency using systems biology approaches. Beyond defining a high-fidelity platform for advancing biPSC-based biotechnologies that have not been previously practicable, these findings also represent a significant step towards understanding corollaries and divergent aspects of bovine pluripotency. This article has an associated First Person interview with the joint first authors of the paper. Summary: Pluripotency reprogramming by overcoming the stable epigenome of bovine cells, and uncovering precise early embryo self-renewal mechanisms enables sustenance and expansion of authentic induced pluripotent stem cells in vitro.
Collapse
Affiliation(s)
- Viju Vijayan Pillai
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Prasanthi P Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Tiffany G Kei
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Shailesh Gurung
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - W Ronald Butler
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY14853, USA
| |
Collapse
|
21
|
Loo CKC, Pearen MA, Ramm GA. The Role of Sonic Hedgehog in Human Holoprosencephaly and Short-Rib Polydactyly Syndromes. Int J Mol Sci 2021; 22:ijms22189854. [PMID: 34576017 PMCID: PMC8468456 DOI: 10.3390/ijms22189854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/03/2021] [Accepted: 09/09/2021] [Indexed: 12/18/2022] Open
Abstract
The Hedgehog (HH) signalling pathway is one of the major pathways controlling cell differentiation and proliferation during human development. This pathway is complex, with HH function influenced by inhibitors, promotors, interactions with other signalling pathways, and non-genetic and cellular factors. Many aspects of this pathway are not yet clarified. The main features of Sonic Hedgehog (SHH) signalling are discussed in relation to its function in human development. The possible role of SHH will be considered using examples of holoprosencephaly and short-rib polydactyly (SRP) syndromes. In these syndromes, there is wide variability in phenotype even with the same genetic mutation, so that other factors must influence the outcome. SHH mutations were the first identified genetic causes of holoprosencephaly, but many other genes and environmental factors can cause malformations in the holoprosencephaly spectrum. Many patients with SRP have genetic defects affecting primary cilia, structures found on most mammalian cells which are thought to be necessary for canonical HH signal transduction. Although SHH signalling is affected in both these genetic conditions, there is little overlap in phenotype. Possible explanations will be canvassed, using data from published human and animal studies. Implications for the understanding of SHH signalling in humans will be discussed.
Collapse
Affiliation(s)
- Christine K. C. Loo
- South Eastern Area Laboratory Services, Department of Anatomical Pathology, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW 2031, Australia
- Correspondence: ; Tel.: +61-2-93829015
| | - Michael A. Pearen
- Hepatic Fibrosis Group, Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (M.A.P.); (G.A.R.)
| | - Grant A. Ramm
- Hepatic Fibrosis Group, Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (M.A.P.); (G.A.R.)
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| |
Collapse
|
22
|
Mecklenburg N, Kowalczyk I, Witte F, Görne J, Laier A, Mamo TM, Gonschior H, Lehmann M, Richter M, Sporbert A, Purfürst B, Hübner N, Hammes A. Identification of disease-relevant modulators of the SHH pathway in the developing brain. Development 2021; 148:272000. [PMID: 34463328 DOI: 10.1242/dev.199307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Pathogenic gene variants in humans that affect the sonic hedgehog (SHH) pathway lead to severe brain malformations with variable penetrance due to unknown modifier genes. To identify such modifiers, we established novel congenic mouse models. LRP2-deficient C57BL/6N mice suffer from heart outflow tract defects and holoprosencephaly caused by impaired SHH activity. These defects are fully rescued on a FVB/N background, indicating a strong influence of modifier genes. Applying comparative transcriptomics, we identified Pttg1 and Ulk4 as candidate modifiers upregulated in the rescue strain. Functional analyses showed that ULK4 and PTTG1, both microtubule-associated proteins, are positive regulators of SHH signaling, rendering the pathway more resilient to disturbances. In addition, we characterized ULK4 and PTTG1 as previously unidentified components of primary cilia in the neuroepithelium. The identification of genes that powerfully modulate the penetrance of genetic disturbances affecting the brain and heart is likely relevant to understanding the variability in human congenital disorders.
Collapse
Affiliation(s)
- Nora Mecklenburg
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Izabela Kowalczyk
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Franziska Witte
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Alena Laier
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Tamrat M Mamo
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Hannes Gonschior
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Martin Lehmann
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Matthias Richter
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Bettina Purfürst
- Electron microscopy technology platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany.,Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Annette Hammes
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| |
Collapse
|
23
|
Arrigo AB, Lin JHI. Endocytic Protein Defects in the Neural Crest Cell Lineage and Its Pathway Are Associated with Congenital Heart Defects. Int J Mol Sci 2021; 22:8816. [PMID: 34445520 PMCID: PMC8396181 DOI: 10.3390/ijms22168816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
Endocytic trafficking is an under-appreciated pathway in cardiac development. Several genes related to endocytic trafficking have been uncovered in a mutagenic ENU screen, in which mutations led to congenital heart defects (CHDs). In this article, we review the relationship between these genes (including LRP1 and LRP2) and cardiac neural crest cells (CNCCs) during cardiac development. Mice with an ENU-induced Lrp1 mutation exhibit a spectrum of CHDs. Conditional deletion using a floxed Lrp1 allele with different Cre drivers showed that targeting neural crest cells with Wnt1-Cre expression replicated the full cardiac phenotypes of the ENU-induced Lrp1 mutation. In addition, LRP1 function in CNCCs is required for normal OFT lengthening and survival/expansion of the cushion mesenchyme, with other cell lineages along the NCC migratory path playing an additional role. Mice with an ENU-induced and targeted Lrp2 mutation demonstrated the cardiac phenotype of common arterial trunk (CAT). Although there is no impact on CNCCs in Lrp2 mutants, the loss of LRP2 results in the depletion of sonic hedgehog (SHH)-dependent cells in the second heart field. SHH is known to be crucial for CNCC survival and proliferation, which suggests LRP2 has a non-autonomous role in CNCCs. In this article, other endocytic trafficking proteins that are associated with CHDs that may play roles in the NCC pathway during development, such as AP1B1, AP2B1, FUZ, MYH10, and HECTD1, are reviewed.
Collapse
Affiliation(s)
- Angelo B. Arrigo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
| | - Jiuann-Huey Ivy Lin
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
24
|
You J, Cheng Y, Yang XJ, Chen L. Generation of a homozygous LRP2 knockout human embryonic stem cell line (FDCHDPe010-A-56) by CRISPR/Cas9 system. Stem Cell Res 2021; 53:102342. [PMID: 33878707 PMCID: PMC9303849 DOI: 10.1016/j.scr.2021.102342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/24/2021] [Accepted: 04/03/2021] [Indexed: 12/15/2022] Open
Abstract
LRP2 is mainly expressed in the cell membrane of epithelia, maintaining normal endocytosis of nutrients from the extracellular microenvironment and mediating growth factor signals. The deficiency of LRP2 can result in abnormal lysosomal and mitochondrial function as well as insufficient resistance to oxidative stress. LRP2-KO animals show enlarged eyes and malfunction of the retinal pigment epithelium (RPE). We were able to generate an LRP2-KO human embryonic stem (ES) cell line using CRISPR/Cas9 gene editing and differentiate the mutant ES cells into RPE cells. Thus, this LRP2-KO human ES line will facilitate studying cellular mechanisms of eye disease due to LRP2 deficiency.
Collapse
Affiliation(s)
- Jie You
- Department of Ophthalmology & Vision Science, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Yun Cheng
- Department of Ophthalmology & Vision Science, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xian-Jie Yang
- Jules Stein Eye Institute, University of California Los Angeles, CA, USA
| | - Ling Chen
- Department of Ophthalmology & Vision Science, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Shanghai Medical School, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Zhou H, Ma Z, Wang Z, Yan S, Wang D, Shen J. Hedgehog signaling regulates regenerative patterning and growth in Harmonia axyridis leg. Cell Mol Life Sci 2021; 78:2185-2197. [PMID: 32909120 PMCID: PMC11071721 DOI: 10.1007/s00018-020-03631-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/07/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
Appendage regeneration has been widely studied in many species. Compared to other animal models, Harmonia axyridis has the advantage of a short life cycle, is easily reared, has strong regeneration capacity and contains systemic RNAi, making it a model organism for research on appendage regeneration. Here, we performed transcriptome analysis, followed by gene functional assays to reveal the molecular mechanism of H. axyridis leg regenerative growth process. Signaling pathways including Decapentaplegic (Dpp), Wingless (Wg), Ds/Ft/Hippo, Notch, Egfr, and Hedgehog (Hh) were all upregulated during the leg regenerative patterning and growth. Among these, Hh and its auxiliary receptor Lrp2 were required for the proper patterning and growth of the regenerative leg. The targets of canonical Hh signaling were required for the regenerative growth which contributes to the leg length, but were not essential for the pattern formation of the regenerative leg. dpp, wg and leg developmental-related genes including rn, dac and Dll were all regulated by hh and lrp2 and may play an essential role in the regenerative patterning of the leg.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China
| | - Zhongzheng Ma
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China
| | - Zhiqi Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China
| | - Shuo Yan
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China.
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
26
|
Kowalczyk I, Lee C, Schuster E, Hoeren J, Trivigno V, Riedel L, Görne J, Wallingford JB, Hammes A, Feistel K. Neural tube closure requires the endocytic receptor Lrp2 and its functional interaction with intracellular scaffolds. Development 2021; 148:dev195008. [PMID: 33500317 PMCID: PMC7860117 DOI: 10.1242/dev.195008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Pathogenic mutations in the endocytic receptor LRP2 in humans are associated with severe neural tube closure defects (NTDs) such as anencephaly and spina bifida. Here, we have combined analysis of neural tube closure in mouse and in the African Clawed Frog Xenopus laevis to elucidate the etiology of Lrp2-related NTDs. Lrp2 loss of function impaired neuroepithelial morphogenesis, culminating in NTDs that impeded anterior neural plate folding and neural tube closure in both model organisms. Loss of Lrp2 severely affected apical constriction as well as proper localization of the core planar cell polarity (PCP) protein Vangl2, demonstrating a highly conserved role of the receptor in these processes, which are essential for neural tube formation. In addition, we identified a novel functional interaction of Lrp2 with the intracellular adaptor proteins Shroom3 and Gipc1 in the developing forebrain. Our data suggest that, during neurulation, motifs within the intracellular domain of Lrp2 function as a hub that orchestrates endocytic membrane removal for efficient apical constriction, as well as PCP component trafficking in a temporospatial manner.
Collapse
Affiliation(s)
- Izabela Kowalczyk
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Chanjae Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Elisabeth Schuster
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Josefine Hoeren
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Valentina Trivigno
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Levin Riedel
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - John B Wallingford
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Annette Hammes
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Kerstin Feistel
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| |
Collapse
|
27
|
Diaz C, Puelles L. Developmental Genes and Malformations in the Hypothalamus. Front Neuroanat 2020; 14:607111. [PMID: 33324176 PMCID: PMC7726113 DOI: 10.3389/fnana.2020.607111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus is a heterogeneous rostral forebrain region that regulates physiological processes essential for survival, energy metabolism, and reproduction, mainly mediated by the pituitary gland. In the updated prosomeric model, the hypothalamus represents the rostralmost forebrain, composed of two segmental regions (terminal and peduncular hypothalamus), which extend respectively into the non-evaginated preoptic telencephalon and the evaginated pallio-subpallial telencephalon. Complex genetic cascades of transcription factors and signaling molecules rule their development. Alterations of some of these molecular mechanisms acting during forebrain development are associated with more or less severe hypothalamic and pituitary dysfunctions, which may be associated with brain malformations such as holoprosencephaly or septo-optic dysplasia. Studies on transgenic mice with mutated genes encoding critical transcription factors implicated in hypothalamic-pituitary development are contributing to understanding the high clinical complexity of these pathologies. In this review article, we will analyze first the complex molecular genoarchitecture of the hypothalamus resulting from the activity of previous morphogenetic signaling centers and secondly some malformations related to alterations in genes implicated in the development of the hypothalamus.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| |
Collapse
|
28
|
Christ A, Marczenke M, Willnow TE. LRP2 controls sonic hedgehog-dependent differentiation of cardiac progenitor cells during outflow tract formation. Hum Mol Genet 2020; 29:3183-3196. [PMID: 32901292 PMCID: PMC7689296 DOI: 10.1093/hmg/ddaa200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Conotruncal malformations are a major cause of congenital heart defects in newborn infants. Recently, genetic screens in humans and in mouse models have identified mutations in LRP2, a multi-ligand receptor, as a novel cause of a common arterial trunk, a severe form of outflow tract (OFT) defect. Yet, the underlying mechanism why the morphogen receptor LRP2 is essential for OFT development remained unexplained. Studying LRP2-deficient mouse models, we now show that LRP2 is expressed in the cardiac progenitor niche of the anterior second heart field (SHF) that contributes to the elongation of the OFT during separation into aorta and pulmonary trunk. Loss of LRP2 in mutant mice results in the depletion of a pool of sonic hedgehog-dependent progenitor cells in the anterior SHF due to premature differentiation into cardiomyocytes as they migrate into the OFT myocardium. Depletion of this cardiac progenitor cell pool results in aberrant shortening of the OFT, the likely cause of CAT formation in affected mice. Our findings identified the molecular mechanism whereby LRP2 controls the maintenance of progenitor cell fate in the anterior SHF essential for OFT separation, and why receptor dysfunction is a novel cause of conotruncal malformation.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Maike Marczenke
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
29
|
Caruso FP, Garofano L, D'Angelo F, Yu K, Tang F, Yuan J, Zhang J, Cerulo L, Pagnotta SM, Bedognetti D, Sims PA, Suvà M, Su XD, Lasorella A, Iavarone A, Ceccarelli M. A map of tumor-host interactions in glioma at single-cell resolution. Gigascience 2020; 9:giaa109. [PMID: 33155039 PMCID: PMC7645027 DOI: 10.1093/gigascience/giaa109] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/08/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Single-cell RNA sequencing is the reference technique for characterizing the heterogeneity of the tumor microenvironment. The composition of the various cell types making up the microenvironment can significantly affect the way in which the immune system activates cancer rejection mechanisms. Understanding the cross-talk signals between immune cells and cancer cells is of fundamental importance for the identification of immuno-oncology therapeutic targets. RESULTS We present a novel method, single-cell Tumor-Host Interaction tool (scTHI), to identify significantly activated ligand-receptor interactions across clusters of cells from single-cell RNA sequencing data. We apply our approach to uncover the ligand-receptor interactions in glioma using 6 publicly available human glioma datasets encompassing 57,060 gene expression profiles from 71 patients. By leveraging this large-scale collection we show that unexpected cross-talk partners are highly conserved across different datasets in the majority of the tumor samples. This suggests that shared cross-talk mechanisms exist in glioma. CONCLUSIONS Our results provide a complete map of the active tumor-host interaction pairs in glioma that can be therapeutically exploited to reduce the immunosuppressive action of the microenvironment in brain tumor.
Collapse
Affiliation(s)
- Francesca Pia Caruso
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
| | - Luciano Garofano
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Fulvio D'Angelo
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Kai Yu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Jinzhou Yuan
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
- Cancer Program, Sidra Medicine, Al Luqta Street, Zone 52, Education City, 26999, Doha Qatar
| | - Jing Zhang
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Luigi Cerulo
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
| | - Stefano M Pagnotta
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
| | - Davide Bedognetti
- Cancer Program, Sidra Medicine, Al Luqta Street, Zone 52, Education City, 26999, Doha Qatar
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Viale Benedetto XV 10, 16132 Genoa, Italy
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 St Nicholas Ave, New York , NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Mario Suvà
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
- Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
| | - Xiao-Dong Su
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, 1130 St Nicholas Ave, New York , NY 10032 USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, 1130 St Nicholas Ave, New York , NY 10032 USA
- Department of Neurology, Columbia University Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
| |
Collapse
|
30
|
Hong M, Christ A, Christa A, Willnow TE, Krauss RS. Cdon mutation and fetal alcohol converge on Nodal signaling in a mouse model of holoprosencephaly. eLife 2020; 9:60351. [PMID: 32876567 PMCID: PMC7467722 DOI: 10.7554/elife.60351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
Holoprosencephaly (HPE), a defect in midline patterning of the forebrain and midface, arises ~1 in 250 conceptions. It is associated with predisposing mutations in the Nodal and Hedgehog (HH) pathways, with penetrance and expressivity graded by genetic and environmental modifiers, via poorly understood mechanisms. CDON is a multifunctional co-receptor, including for the HH pathway. In mice, Cdon mutation synergizes with fetal alcohol exposure, producing HPE phenotypes closely resembling those seen in humans. We report here that, unexpectedly, Nodal signaling is a major point of synergistic interaction between Cdon mutation and fetal alcohol. Window-of-sensitivity, genetic, and in vitro findings are consistent with a model whereby brief exposure of Cdon mutant embryos to ethanol during gastrulation transiently and partially inhibits Nodal pathway activity, with consequent effects on midline patterning. These results illuminate mechanisms of gene-environment interaction in a multifactorial model of a common birth defect.
Collapse
Affiliation(s)
- Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Annabel Christ
- Max-Delbruck-Center for Molecular Medicine, Berlin, Germany
| | - Anna Christa
- Max-Delbruck-Center for Molecular Medicine, Berlin, Germany
| | | | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
31
|
Induced pluripotent stem cell-based disease modeling identifies ligand-induced decay of megalin as a cause of Donnai-Barrow syndrome. Kidney Int 2020; 98:159-167. [PMID: 32471643 PMCID: PMC7322522 DOI: 10.1016/j.kint.2020.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 01/16/2023]
Abstract
Donnai-Barrow syndrome (DBS) is an autosomal-recessive disorder characterized by multiple pathologies including malformation of forebrain and eyes, as well as resorption defects of the kidney proximal tubule. The underlying cause of DBS are mutations in LRP2, encoding the multifunctional endocytic receptor megalin. Here, we identified a unique missense mutation R3192Q of LRP2 in an affected family that may provide novel insights into the molecular causes of receptor dysfunction in the kidney proximal tubule and other tissues affected in DBS. Using patient-derived induced pluripotent stem cell lines we generated neuroepithelial and kidney cell types as models of the disease. Using these cell models, we documented the inability of megalin R3192Q to properly discharge ligand and ligand-induced receptor decay in lysosomes. Thus, mutant receptors are aberrantly targeted to lysosomes for catabolism, essentially depleting megalin in the presence of ligand in this affected family.
Collapse
|
32
|
Single-Cell Transcriptomics Characterizes Cell Types in the Subventricular Zone and Uncovers Molecular Defects Impairing Adult Neurogenesis. Cell Rep 2019; 25:2457-2469.e8. [PMID: 30485812 DOI: 10.1016/j.celrep.2018.11.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/13/2018] [Accepted: 10/31/2018] [Indexed: 12/26/2022] Open
Abstract
Neural stem cells (NSCs) contribute to plasticity and repair of the adult brain. Niches harboring NSCs regulate stem cell self-renewal and differentiation. We used comprehensive and untargeted single-cell RNA profiling to generate a molecular cell atlas of the largest germinal region of the adult mouse brain, the subventricular zone (SVZ). We characterized >20 neural and non-neural cell types and gained insights into the dynamics of neurogenesis by predicting future cell states based on computational analysis of RNA kinetics. Furthermore, we applied our single-cell approach to document decreased numbers of NSCs, reduced proliferation activity of progenitors, and perturbations in Wnt and BMP signaling pathways in mice lacking LRP2, an endocytic receptor required for SVZ maintenance. Our data provide a valuable resource to study adult neurogenesis and a proof of principle for the power of single-cell RNA sequencing to elucidate neural cell-type-specific alterations in loss-of-function models.
Collapse
|
33
|
Sasai N, Toriyama M, Kondo T. Hedgehog Signal and Genetic Disorders. Front Genet 2019; 10:1103. [PMID: 31781166 PMCID: PMC6856222 DOI: 10.3389/fgene.2019.01103] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The hedgehog (Hh) family comprises sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh), which are versatile signaling molecules involved in a wide spectrum of biological events including cell differentiation, proliferation, and survival; establishment of the vertebrate body plan; and aging. These molecules play critical roles from embryogenesis to adult stages; therefore, alterations such as abnormal expression or mutations of the genes involved and their downstream factors cause a variety of genetic disorders at different stages. The Hh family involves many signaling mediators and functions through complex mechanisms, and achieving a comprehensive understanding of the entire signaling system is challenging. This review discusses the signaling mediators of the Hh pathway and their functions at the cellular and organismal levels. We first focus on the roles of Hh signaling mediators in signal transduction at the cellular level and the networks formed by these factors. Then, we analyze the spatiotemporal pattern of expression of Hh pathway molecules in tissues and organs, and describe the phenotypes of mutant mice. Finally, we discuss the genetic disorders caused by malfunction of Hh signaling-related molecules in humans.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Systems Neurobiology and Medicine, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
34
|
Kim A, Savary C, Dubourg C, Carré W, Mouden C, Hamdi-Rozé H, Guyodo H, Douce JL, Pasquier L, Flori E, Gonzales M, Bénéteau C, Boute O, Attié-Bitach T, Roume J, Goujon L, Akloul L, Odent S, Watrin E, Dupé V, de Tayrac M, David V. Integrated clinical and omics approach to rare diseases: novel genes and oligogenic inheritance in holoprosencephaly. Brain 2019; 142:35-49. [PMID: 30508070 DOI: 10.1093/brain/awy290] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022] Open
Abstract
Holoprosencephaly is a pathology of forebrain development characterized by high phenotypic heterogeneity. The disease presents with various clinical manifestations at the cerebral or facial levels. Several genes have been implicated in holoprosencephaly but its genetic basis remains unclear: different transmission patterns have been described including autosomal dominant, recessive and digenic inheritance. Conventional molecular testing approaches result in a very low diagnostic yield and most cases remain unsolved. In our study, we address the possibility that genetically unsolved cases of holoprosencephaly present an oligogenic origin and result from combined inherited mutations in several genes. Twenty-six unrelated families, for whom no genetic cause of holoprosencephaly could be identified in clinical settings [whole exome sequencing and comparative genomic hybridization (CGH)-array analyses], were reanalysed under the hypothesis of oligogenic inheritance. Standard variant analysis was improved with a gene prioritization strategy based on clinical ontologies and gene co-expression networks. Clinical phenotyping and exploration of cross-species similarities were further performed on a family-by-family basis. Statistical validation was performed on 248 ancestrally similar control trios provided by the Genome of the Netherlands project and on 574 ancestrally matched controls provided by the French Exome Project. Variants of clinical interest were identified in 180 genes significantly associated with key pathways of forebrain development including sonic hedgehog (SHH) and primary cilia. Oligogenic events were observed in 10 families and involved both known and novel holoprosencephaly genes including recurrently mutated FAT1, NDST1, COL2A1 and SCUBE2. The incidence of oligogenic combinations was significantly higher in holoprosencephaly patients compared to two control populations (P < 10-9). We also show that depending on the affected genes, patients present with particular clinical features. This study reports novel disease genes and supports oligogenicity as clinically relevant model in holoprosencephaly. It also highlights key roles of SHH signalling and primary cilia in forebrain development. We hypothesize that distinction between different clinical manifestations of holoprosencephaly lies in the degree of overall functional impact on SHH signalling. Finally, we underline that integrating clinical phenotyping in genetic studies is a powerful tool to specify the clinical relevance of certain mutations.
Collapse
Affiliation(s)
- Artem Kim
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Clara Savary
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Christèle Dubourg
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Wilfrid Carré
- Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Charlotte Mouden
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Houda Hamdi-Rozé
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Hélène Guyodo
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Jerome Le Douce
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | | | | | | | - Elisabeth Flori
- Laboratoire de Cytogénétique, Cytologie et Histologie Quantitative, Hôpital de Hautepierre, HUS, Strasbourg, France
| | - Marie Gonzales
- Service de Génétique et Embryologie Médicales, Hôpital Armand Trousseau, Paris, France
| | | | | | - Tania Attié-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants-Malades, Université Paris Descartes, 149, rue de Sèvres, Paris, France
| | - Joelle Roume
- Department of Clinical Genetics, Centre de Référence “AnDDI Rares”, Poissy Hospital GHU PIFO, Poissy, France
| | | | - Linda Akloul
- Service de Génétique Clinique, CHU, Rennes, France
| | - Sylvie Odent
- Service de Génétique Clinique, CHU, Rennes, France
| | - Erwan Watrin
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Valérie Dupé
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Marie de Tayrac
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Véronique David
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| |
Collapse
|
35
|
Renard E, Chéry C, Oussalah A, Josse T, Perrin P, Tramoy D, Voirin J, Klein O, Leheup B, Feillet F, Guéant-Rodriguez RM, Guéant JL. Exome sequencing of cases with neural tube defects identifies candidate genes involved in one-carbon/vitamin B12 metabolisms and Sonic Hedgehog pathway. Hum Genet 2019; 138:703-713. [DOI: 10.1007/s00439-019-02015-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/13/2019] [Indexed: 12/13/2022]
|
36
|
Carter SP, Blacque OE. Membrane retrieval, recycling and release pathways that organise and sculpt the ciliary membrane. Curr Opin Cell Biol 2019; 59:133-139. [PMID: 31146146 DOI: 10.1016/j.ceb.2019.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/16/2022]
Abstract
The microtubule-based cilium that extends from the surface of most eukaryotic cell types serves motility, sensory reception and cell-cell signaling functions, and is disrupted in wide-ranging ciliopathy disorders. The cilium is heavily reliant on dynamic and tuneable intracellular trafficking systems such as intraflagellar transport and Golgi-derived secretory pathways, which control the organelle's structure, function and molecular composition. More recently, endosomal retrieval and recycling, as well as extracellular vesicle (EV) release, pathways have been associated with ciliary membrane control. Here, we discuss the emerging role of these pathways in the control of ciliary membrane homeostasis. The new findings provide a deeper and more integrated understanding of how the ciliary membrane is organised.
Collapse
Affiliation(s)
- Stephen P Carter
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
37
|
Ferent J, Giguère F, Jolicoeur C, Morin S, Michaud JF, Makihara S, Yam PT, Cayouette M, Charron F. Boc Acts via Numb as a Shh-Dependent Endocytic Platform for Ptch1 Internalization and Shh-Mediated Axon Guidance. Neuron 2019; 102:1157-1171.e5. [PMID: 31054872 DOI: 10.1016/j.neuron.2019.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/08/2019] [Accepted: 03/28/2019] [Indexed: 01/14/2023]
Abstract
During development, Shh attracts commissural axons toward the floor plate through a non-canonical, transcription-independent signaling pathway that requires the receptor Boc. Here, we find that Shh induces Boc internalization into early endosomes and that endocytosis is required for Shh-mediated growth-cone turning. Numb, an endocytic adaptor, binds to Boc and is required for Boc internalization, Shh-mediated growth-cone turning in vitro, and commissural axon guidance in vivo. Similar to Boc, Ptch1 is also internalized by Shh in a Numb-dependent manner; however, the binding of Shh to Ptch1 alone is not sufficient to induce Ptch1 internalization nor growth-cone turning. Therefore, the binding of Shh to Boc is required for Ptch1 internalization and growth-cone turning. Our data support a model where Boc endocytosis via Numb is required for Ptch1 internalization and Shh signaling in axon guidance. Thus, Boc acts as a Shh-dependent endocytic platform gating Ptch1 internalization and Shh signaling.
Collapse
Affiliation(s)
- Julien Ferent
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Neuroscience, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Fanny Giguère
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Christine Jolicoeur
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Steves Morin
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Jean-Francois Michaud
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Shirin Makihara
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Patricia T Yam
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
| | - Michel Cayouette
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada; Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Frederic Charron
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada; Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada; Department of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; Department of Biology, McGill University, Montreal, QC H3A 0G4, Canada.
| |
Collapse
|
38
|
Abstract
Cell-to-cell communication is fundamental for embryo development and subsequent tissue homeostasis. This communication is often mediated by a small number of signaling pathways in which a secreted ligand binds to the surface of a target cell, thereby activating signal transduction. In vertebrate neural development, these signaling mechanisms are repeatedly used to obtain different and context-dependent outcomes. Part of the versatility of these communication mechanisms depends on their finely tuned regulation that controls timing, spatial localization, and duration of the signaling. The existence of secreted antagonists, which prevent ligand–receptor interaction, is an efficient mechanism to regulate some of these pathways. The Hedgehog family of signaling proteins, however, activates a pathway that is controlled largely by the positive or negative activity of membrane-bound proteins such as Cdon, Boc, Gas1, or Megalin/LRP2. In this review, we will use the development of the vertebrate retina, from its early specification to neurogenesis, to discuss whether there is an advantage to the use of such regulators, pointing to unresolved or controversial issues.
Collapse
Affiliation(s)
- Viviana Gallardo
- Centro de Biología Molecular , CSIC-UAM, Madrid, 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular , CSIC-UAM, Madrid, 28049, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| |
Collapse
|
39
|
Corman TS, Bergendahl SE, Epstein DJ. Distinct temporal requirements for Sonic hedgehog signaling in development of the tuberal hypothalamus. Development 2018; 145:dev167379. [PMID: 30291164 PMCID: PMC6240319 DOI: 10.1242/dev.167379] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022]
Abstract
Sonic hedgehog (Shh) plays well characterized roles in brain and spinal cord development, but its functions in the hypothalamus have been more difficult to elucidate owing to the complex neuroanatomy of this brain area. Here, we use fate mapping and conditional deletion models in mice to define requirements for dynamic Shh activity at distinct developmental stages in the tuberal hypothalamus, a brain region with important homeostatic functions. At early time points, Shh signaling regulates dorsoventral patterning, neurogenesis and the size of the ventral midline. Fate-mapping experiments demonstrate that Shh-expressing and -responsive progenitors contribute to distinct neuronal subtypes, accounting for some of the cellular heterogeneity in tuberal hypothalamic nuclei. Conditional deletion of the hedgehog transducer smoothened (Smo), after dorsoventral patterning has been established, reveals that Shh signaling is necessary to maintain proliferation and progenitor identity during peak periods of hypothalamic neurogenesis. We also find that mosaic disruption of Smo causes a non-cell autonomous gain in Shh signaling activity in neighboring wild-type cells, suggesting a mechanism for the pathogenesis of hypothalamic hamartomas, benign tumors that form during hypothalamic development.
Collapse
Affiliation(s)
- Tanya S Corman
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| | - Solsire E Bergendahl
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| |
Collapse
|
40
|
Dong C, Cui D, Liu G, Xu H, Peng X, Duan J, Liu L. Cancer stem cell associated eight gene-based signature predicts clinical outcomes of colorectal cancer. Oncol Lett 2018; 17:442-449. [PMID: 30655785 DOI: 10.3892/ol.2018.9533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/20/2017] [Indexed: 12/21/2022] Open
Abstract
Previous studies have suggested that cancer stem cells serve crucial functions in tumorigenesis, metastasis and therapy failure. Stem cell signaling transduction pathways are frequently dysregulated in cancer and associated with tumorigenesis, metastasis and the cell cycle, which are necessary for cancer proliferation. However, cancer stem cell-associated gene signatures have not been established for predicting patient outcomes in colorectal cancer. Using a gene-mining approach, the present study performed mRNA expression profiling in large colorectal cancer cohorts from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database, including a TCGA colorectal cancer cohort (n=383) and three independent validation series GSE39582 (n=582), GSE17536 (n=177) and GSE17537 (n=55). The present study identified that an eight-gene signature in cancer stem cell signaling was associated with the overall survival and disease/recurrence-free survival of patients with colorectal. On the basis of this signature, patients in the TCGA training sets were divided into high-risk and low-risk subgroups with a significantly different overall survival rate (hazard ratio, 2.38; P=0.0005). The prognostic value of this signature was confirmed using three independent GEO colorectal cancer sets. Identifying this prognostic stem cell signaling signature may provide an efficient classification tool for clinical prognosis evaluation, and facilitate cancer stem cell-targeted therapy.
Collapse
Affiliation(s)
- Chuanpeng Dong
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Danni Cui
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Gang Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Huilin Xu
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Xueqing Peng
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Juan Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China.,Translational Medicine Institute, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Lei Liu
- Institute of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
41
|
Matías-Guíu J, Oreja-Guevara C, Matias-Guiu J, Gomez-Pinedo U. Vitamin D and remyelination in multiple sclerosis. NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2016.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
42
|
Sabatino JA, Stokes BA, Zohn IE. Prevention of neural tube defects in Lrp2 mutant mouse embryos by folic acid supplementation. Birth Defects Res 2018; 109:16-26. [PMID: 27883261 DOI: 10.1002/bdra.23589] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/18/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Neural tube defects (NTDs) are among the most common structural birth defects in humans and are caused by the complex interaction of genetic and environmental factors. Periconceptional supplementation with folic acid can prevent NTDs in both mouse models and human populations. A better understanding of how genes and environmental factors interact is critical toward development of rational strategies to prevent NTDs. Low density lipoprotein-related protein 2 (Lrp2) is involved in endocytosis of the folic acid receptor among numerous other nutrients and ligands. METHODS We determined the effect of iron and/or folic acid supplementation on the penetrance of NTDs in the Lrp2null mouse model. The effects of supplementation on folate and iron status were measured in embryos and dams. RESULTS Periconceptional dietary supplementation with folic acid did not prevent NTDs in Lrp2 mutant embryos, whereas high levels of folic acid supplementation by intraperitoneal injection reduced incidence of NTDs. Importantly, Lrp2null/+ dams had reduced blood folate levels that improved with daily intraperitoneal injections of folate but not dietary supplementation. On the contrary, iron supplementation had no effect on the penetrance of NTDs in Lrp2 mutant embryos and negated the preventative effect of folic acid supplementation in Lrp2null/null mutants. CONCLUSION Lrp2 is required for folate homeostasis in heterozygous dams and high levels of supplementation prevents NTDs. Furthermore, high levels of dietary iron supplementation interfered with folic acid supplementation negating the positive effects of supplementation in this model. Birth Defects Research 109:16-26, 2017. © 2016 The Authors Birth Defects Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Julia A Sabatino
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Bethany A Stokes
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC.,Department of Biology, The George Washington University, Washington, DC
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| |
Collapse
|
43
|
Abstract
Cilia are microtubule-based organelles extending from a basal body at the surface of eukaryotic cells. Cilia regulate cell and fluid motility, sensation and developmental signaling, and ciliary defects cause human diseases (ciliopathies) affecting the formation and function of many tissues and organs. Over the past decade, various Rab and Rab-like membrane trafficking proteins have been shown to regulate cilia-related processes such as basal body maturation, ciliary axoneme extension, intraflagellar transport and ciliary signaling. In this review, we provide a comprehensive overview of Rab protein ciliary associations, drawing on findings from multiple model systems, including mammalian cell culture, mice, zebrafish, C. elegans, trypanosomes, and green algae. We also discuss several emerging mechanistic themes related to ciliary Rab cascades and functional redundancy.
Collapse
Affiliation(s)
- Oliver E Blacque
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| | - Noemie Scheidel
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| | - Stefanie Kuhns
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| |
Collapse
|
44
|
Wiegering A, Rüther U, Gerhardt C. The Role of Hedgehog Signalling in the Formation of the Ventricular Septum. J Dev Biol 2017; 5:E17. [PMID: 29615572 PMCID: PMC5831794 DOI: 10.3390/jdb5040017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 12/11/2022] Open
Abstract
An incomplete septation of the ventricles in the vertebrate heart that disturbes the strict separation between the contents of the two ventricles is termed a ventricular septal defect (VSD). Together with bicuspid aortic valves, it is the most frequent congenital heart disease in humans. Until now, life-threatening VSDs are usually treated surgically. To avoid surgery and to develop an alternative therapy (e.g., a small molecule therapy), it is necessary to understand the molecular mechanisms underlying ventricular septum (VS) development. Consequently, various studies focus on the investigation of signalling pathways, which play essential roles in the formation of the VS. In the past decade, several reports found evidence for an involvement of Hedgehog (HH) signalling in VS development. In this review article, we will summarise the current knowledge about the association between HH signalling and VS formation and discuss the use of such knowledge to design treatment strategies against the development of VSDs.
Collapse
Affiliation(s)
- Antonia Wiegering
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Ulrich Rüther
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Christoph Gerhardt
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
45
|
Abstract
Cilia are microtubule-based organelles extending from a basal body at the surface of eukaryotic cells. Cilia regulate cell and fluid motility, sensation and developmental signaling, and ciliary defects cause human diseases (ciliopathies) affecting the formation and function of many tissues and organs. Over the past decade, various Rab and Rab-like membrane trafficking proteins have been shown to regulate cilia-related processes such as basal body maturation, ciliary axoneme extension, intraflagellar transport and ciliary signaling. In this review, we provide a comprehensive overview of Rab protein ciliary associations, drawing on findings from multiple model systems, including mammalian cell culture, mice, zebrafish, C. elegans, trypanosomes, and green algae. We also discuss several emerging mechanistic themes related to ciliary Rab cascades and functional redundancy.
Collapse
Affiliation(s)
- Oliver E Blacque
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| | - Noemie Scheidel
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| | - Stefanie Kuhns
- a School of Biomolecular and Biomedical Science , University College Dublin , Belfield, Dublin , Ireland
| |
Collapse
|
46
|
Gene Expression Networks in the Murine Pulmonary Myocardium Provide Insight into the Pathobiology of Atrial Fibrillation. G3-GENES GENOMES GENETICS 2017; 7:2999-3017. [PMID: 28720711 PMCID: PMC5592927 DOI: 10.1534/g3.117.044651] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pulmonary myocardium is a muscular coat surrounding the pulmonary and caval veins. Although its definitive physiological function is unknown, it may have a pathological role as the source of ectopic beats initiating atrial fibrillation. How the pulmonary myocardium gains pacemaker function is not clearly defined, although recent evidence indicates that changed transcriptional gene expression networks are at fault. The gene expression profile of this distinct cell type in situ was examined to investigate underlying molecular events that might contribute to atrial fibrillation. Via systems genetics, a whole-lung transcriptome data set from the BXD recombinant inbred mouse resource was analyzed, uncovering a pulmonary cardiomyocyte gene network of 24 transcripts, coordinately regulated by chromosome 1 and 2 loci. Promoter enrichment analysis and interrogation of publicly available ChIP-seq data suggested that transcription of this gene network may be regulated by the concerted activity of NKX2-5, serum response factor, myocyte enhancer factor 2, and also, at a post-transcriptional level, by RNA binding protein motif 20. Gene ontology terms indicate that this gene network overlaps with molecular markers of the stressed heart. Therefore, we propose that perturbed regulation of this gene network might lead to altered calcium handling, myocyte growth, and contractile force contributing to the aberrant electrophysiological properties observed in atrial fibrillation. We reveal novel molecular interactions and pathways representing possible therapeutic targets for atrial fibrillation. In addition, we highlight the utility of recombinant inbred mouse resources in detecting and characterizing gene expression networks of relatively small populations of cells that have a pathological significance.
Collapse
|
47
|
Yang T, Williams BO. Low-Density Lipoprotein Receptor-Related Proteins in Skeletal Development and Disease. Physiol Rev 2017; 97:1211-1228. [PMID: 28615463 DOI: 10.1152/physrev.00013.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
The identification of the low-density lipoprotein receptor (LDLR) provided a foundation for subsequent studies in lipoprotein metabolism, receptor-mediated endocytosis, and many other fundamental biological functions. The importance of the LDLR led to numerous studies that identified homologous molecules and ultimately resulted in the description of the LDL-receptor superfamily, a group of proteins that contain domains also found in the LDLR. Subsequent studies have revealed that members of the LDLR-related protein family play roles in regulating many aspects of signal transduction. This review is focused on the roles of selected members of this protein family in skeletal development and disease. We present background on the identification of this subgroup of receptors, discuss the phenotypes associated with alterations in their function in human patients and mouse models, and describe the current efforts to therapeutically target these proteins to treat human skeletal disease.
Collapse
Affiliation(s)
- Tao Yang
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Bart O Williams
- Program in Skeletal Disease and Tumor Microenvironment, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|
48
|
Pinskey JM, Franks NE, McMellen AN, Giger RJ, Allen BL. Neuropilin-1 promotes Hedgehog signaling through a novel cytoplasmic motif. J Biol Chem 2017; 292:15192-15204. [PMID: 28667171 DOI: 10.1074/jbc.m117.783845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/23/2017] [Indexed: 12/30/2022] Open
Abstract
Hedgehog (HH) signaling critically regulates embryonic and postnatal development as well as adult tissue homeostasis, and its perturbation can lead to developmental disorders, birth defects, and cancers. Neuropilins (NRPs), which have well-defined roles in Semaphorin and VEGF signaling, positively regulate HH pathway function, although their mechanism of action in HH signaling remains unclear. Here, using luciferase-based reporter assays, we provide evidence that NRP1 regulates HH signaling specifically at the level of GLI transcriptional activator function. Moreover, we show that NRP1 localization to the primary cilium, a key platform for HH signal transduction, does not correlate with HH signal promotion. Rather, a structure-function analysis suggests that the NRP1 cytoplasmic and transmembrane domains are necessary and sufficient to regulate HH pathway activity. Furthermore, we identify a previously uncharacterized, 12-amino acid region within the NRP1 cytoplasmic domain that mediates HH signal promotion. Overall, our results provide mechanistic insight into NRP1 function within and potentially beyond the HH signaling pathway. These insights have implications for the development of novel modulators of HH-driven developmental disorders and diseases.
Collapse
Affiliation(s)
- Justine M Pinskey
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Nicole E Franks
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Alexandra N McMellen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Roman J Giger
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin L Allen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
49
|
Lažetić V, Fay DS. Molting in C. elegans. WORM 2017; 6:e1330246. [PMID: 28702275 DOI: 10.1080/21624054.2017.1330246] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
Molting is an essential developmental process for the majority of animal species on Earth. During the molting process, which is a specialized form of extracellular matrix (ECM) remodeling, the old apical ECM, or cuticle, is replaced with a new one. Many of the genes and pathways identified as important for molting in nematodes are highly conserved in vertebrates and include regulators and components of vesicular trafficking, steroid-hormone signaling, developmental timers, and hedgehog-like signaling. In this review, we discuss what is known about molting, with a focus on studies in Caenorhabditis elegans. We also describe the key structural elements of the cuticle that must be released, newly synthesized, or remodeled for proper molting to occur.
Collapse
Affiliation(s)
- Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, USA
| | - David S Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
50
|
Endocytic receptor LRP2/megalin—of holoprosencephaly and renal Fanconi syndrome. Pflugers Arch 2017; 469:907-916. [DOI: 10.1007/s00424-017-1992-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 12/31/2022]
|