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
|
Yu X, Wang X, Ma K, Gao D, Deng Y, Zhou D, Ding W, Zhao Y, Liu Q, Zhou Z. Tai/NCOA2 suppresses the Hedgehog pathway by directly targeting the transcription factor Ci/GLI. Proc Natl Acad Sci U S A 2024; 121:e2409380121. [PMID: 39531503 PMCID: PMC11588115 DOI: 10.1073/pnas.2409380121] [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: 05/12/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
The Hedgehog (Hh) pathway plays diverse roles in cellular processes by activating the transcription factor Cubitus interruptus (Ci). Abnormal regulation of this pathway has been linked to various human diseases. While previous studies have focused on how Ci is regulated in the cytoplasm, the control of nuclear Ci remains poorly understood. In this study, we have found that the transcriptional cofactor Taiman (Tai) functions as an inhibitor of the Hh pathway. Tai interferes with the response of Hh signal, rather than Hh secretion. Our epistatic analyses reveal that Tai works in parallel with Ci to reduce its activity, thereby counteracting organ overgrowth and the activation of target genes caused by Ci overexpression. Specifically, Tai interacts with Ci to decrease its binding to target gene promoters. The Hh signal weakens the interaction between Ci and Tai, releasing the inhibition on Ci. Importantly, this regulatory mechanism is conserved from Drosophila to mammalian cells. Moreover, NCOA1-3 are the mammalian ortholog of Drosophila protein Tai, but only NCOA2 plays a similar role in inhibiting the Hh pathway. These findings reveal an additional way to modulate the transcriptional activity of nuclear Ci.
Collapse
Affiliation(s)
- Xuan Yu
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Xinyu Wang
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Kaize Ma
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Dongqing Gao
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Yanran Deng
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang330022, China
| | - Dafa Zhou
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Wenhao Ding
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Yunhe Zhao
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Qingxin Liu
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
| | - Zizhang Zhou
- College of Life Sciences, Shandong Agricultural University, Tai’an271018, China
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang330022, China
| |
Collapse
|
3
|
Konopka A, Gawin K, Barszcz M. Hedgehog Signalling Pathway and Its Role in Shaping the Architecture of Intestinal Epithelium. Int J Mol Sci 2024; 25:12007. [PMID: 39596072 PMCID: PMC11593361 DOI: 10.3390/ijms252212007] [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: 10/07/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The hedgehog (Hh) signalling pathway plays a key role in both embryonic and postnatal development of the intestine and is responsible for gut homeostasis. It regulates stem cell renewal, formation of the villous-crypt axis, differentiation of goblet and Paneth cells, the cell cycle, apoptosis, development of gut innervation, and lipid metabolism. Ligands of the Hh pathway, i.e., Indian hedgehog (Ihh) and Sonic hedgehog (Shh), are expressed by superficial enterocytes but act in the mesenchyme, where they are bound by a Patched receptor localised on myofibroblasts and smooth muscle cells. This activates a cascade leading to the transcription of target genes, including those encoding G1/S-specific cyclin-D2 and -E1, B-cell lymphoma 2, fibroblast growth factor 4, and bone morphogenetic protein 4. The Hh pathway is tightly connected to Wnt signalling. Ihh is the major ligand in the Hh pathway. Its activation inhibits proliferation, while its blocking induces hyperproliferation and triggers a wound-healing response. Thus, Ihh is a negative feedback regulator of cell proliferation. There are data indicating that diet composition may affect the expression of the Hh pathway genes and proteins, which in turn, induces changes in mucosal architecture. This was shown for fat, vitamin A, haem, berberine, and ovotransferrin. The Hh signalling is also affected by the intestinal microbiota, which affects the intestinal barrier integrity. This review highlights the critical importance of the Hh pathway in shaping the intestinal mucosa and summarises the results obtained so far in research on the effect of dietary constituents on the activity of this pathway.
Collapse
Affiliation(s)
- Adrianna Konopka
- Laboratory of Analysis of Gastrointestinal Tract Protective Barrier, Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Kamil Gawin
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Marcin Barszcz
- Laboratory of Analysis of Gastrointestinal Tract Protective Barrier, Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| |
Collapse
|
4
|
Ehring K, Ehlers SF, Froese J, Gude F, Puschmann J, Grobe K. Two-way Dispatched function in Sonic hedgehog shedding and transfer to high-density lipoproteins. eLife 2024; 12:RP86920. [PMID: 39297609 PMCID: PMC11412720 DOI: 10.7554/elife.86920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
The Sonic hedgehog (Shh) signaling pathway controls embryonic development and tissue homeostasis after birth. This requires regulated solubilization of dual-lipidated, firmly plasma membrane-associated Shh precursors from producing cells. Although it is firmly established that the resistance-nodulation-division transporter Dispatched (Disp) drives this process, it is less clear how lipidated Shh solubilization from the plasma membrane is achieved. We have previously shown that Disp promotes proteolytic solubilization of Shh from its lipidated terminal peptide anchors. This process, termed shedding, converts tightly membrane-associated hydrophobic Shh precursors into delipidated soluble proteins. We show here that Disp-mediated Shh shedding is modulated by a serum factor that we identify as high-density lipoprotein (HDL). In addition to serving as a soluble sink for free membrane cholesterol, HDLs also accept the cholesterol-modified Shh peptide from Disp. The cholesteroylated Shh peptide is necessary and sufficient for Disp-mediated transfer because artificially cholesteroylated mCherry associates with HDL in a Disp-dependent manner, whereas an N-palmitoylated Shh variant lacking C-cholesterol does not. Disp-mediated Shh transfer to HDL is completed by proteolytic processing of the palmitoylated N-terminal membrane anchor. In contrast to dual-processed soluble Shh with moderate bioactivity, HDL-associated N-processed Shh is highly bioactive. We propose that the purpose of generating different soluble forms of Shh from the dual-lipidated precursor is to tune cellular responses in a tissue-type and time-specific manner.
Collapse
Affiliation(s)
- Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | | | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Janna Puschmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| |
Collapse
|
5
|
Schlissel G, Meziane M, Narducci D, Hansen AS, Li P. Diffusion barriers imposed by tissue topology shape Hedgehog morphogen gradients. Proc Natl Acad Sci U S A 2024; 121:e2400677121. [PMID: 39190357 PMCID: PMC11388384 DOI: 10.1073/pnas.2400677121] [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: 01/11/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
Animals use a small number of morphogens to pattern tissues, but it is unclear how evolution modulates morphogen signaling range to match tissues of varying sizes. Here, we used single-molecule imaging in reconstituted morphogen gradients and in tissue explants to determine that Hedgehog diffused extracellularly as a monomer, and rapidly transitioned between membrane-confined and -unconfined states. Unexpectedly, the vertebrate-specific protein SCUBE1 expanded Hedgehog gradients by accelerating the transition rates between states without affecting the relative abundance of molecules in each state. This observation could not be explained under existing models of morphogen diffusion. Instead, we developed a topology-limited diffusion model in which cell-cell gaps create diffusion barriers, which morphogens can only overcome by passing through a membrane-unconfined state. Under this model, SCUBE1 promoted Hedgehog secretion and diffusion by allowing it to transiently overcome diffusion barriers. This multiscale understanding of morphogen gradient formation unified prior models and identified knobs that nature can use to tune morphogen gradient sizes across tissues and organisms.
Collapse
Affiliation(s)
- Gavin Schlissel
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | - Miram Meziane
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Domenic Narducci
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA02142
- Koch Institute for Integrative Cancer Research, Cambridge, MA02139
| | - Anders S. Hansen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA02142
- Koch Institute for Integrative Cancer Research, Cambridge, MA02139
| | - Pulin Li
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
6
|
Lavillaureix A, Rollier P, Kim A, Panasenkava V, De Tayrac M, Carré W, Guyodo H, Faoucher M, Poirel E, Akloul L, Quélin C, Whalen S, Bos J, Broekema M, van Hagen JM, Grand K, Allen-Sharpley M, Magness E, McLean SD, Kayserili H, Altunoglu U, En Qi Chong A, Xue S, Jeanne M, Almontashiri N, Habhab W, Vanlerberghe C, Faivre L, Viora-Dupont E, Philippe C, Safraou H, Laffargue F, Mittendorf L, Abou Jamra R, Patil SJ, Dalal A, Sarma AS, Keren B, Reversade B, Dubourg C, Odent S, Dupé V. DISP1 deficiency: Monoallelic and biallelic variants cause a spectrum of midline craniofacial malformations. Genet Med 2024; 26:101126. [PMID: 38529886 DOI: 10.1016/j.gim.2024.101126] [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: 06/07/2023] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
PURPOSE DISP1 encodes a transmembrane protein that regulates the secretion of the morphogen, Sonic hedgehog, a deficiency of which is a major cause of holoprosencephaly (HPE). This disorder covers a spectrum of brain and midline craniofacial malformations. The objective of the present study was to better delineate the clinical phenotypes associated with division transporter dispatched-1 (DISP1) variants. METHODS This study was based on the identification of at least 1 pathogenic variant of the DISP1 gene in individuals for whom detailed clinical data were available. RESULTS A total of 23 DISP1 variants were identified in heterozygous, compound heterozygous or homozygous states in 25 individuals with midline craniofacial defects. Most cases were minor forms of HPE, with craniofacial features such as orofacial cleft, solitary median maxillary central incisor, and congenital nasal pyriform aperture stenosis. These individuals had either monoallelic loss-of-function variants or biallelic missense variants in DISP1. In individuals with severe HPE, the DISP1 variants were commonly found associated with a variant in another HPE-linked gene (ie, oligogenic inheritance). CONCLUSION The genetic findings we have acquired demonstrate a significant involvement of DISP1 variants in the phenotypic spectrum of midline defects. This underlines its importance as a crucial element in the efficient secretion of Sonic hedgehog. We also demonstrated that the very rare solitary median maxillary central incisor and congenital nasal pyriform aperture stenosis combination is part of the DISP1-related phenotype. The present study highlights the clinical risks to be flagged up during genetic counseling after the discovery of a pathogenic DISP1 variant.
Collapse
Affiliation(s)
- Alinoë Lavillaureix
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France; Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France
| | - Paul Rollier
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France; Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France
| | - Artem Kim
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France; Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Veranika Panasenkava
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France
| | - Marie De Tayrac
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France; Génétique Moléculaire et Génomique, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Wilfrid Carré
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France; Génétique Moléculaire et Génomique, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Hélène Guyodo
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France
| | - Marie Faoucher
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France; Génétique Moléculaire et Génomique, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Elisabeth Poirel
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Linda Akloul
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Chloé Quélin
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Sandra Whalen
- APHP, Sorbonne Université, Département de Génétique, Centre de Référence Maladies Rares des Anomalies du Développement et Syndromes Malformatifs, Hôpital Trousseau & Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jessica Bos
- Department of Human Genetics, Section Clinical Genetic, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marjoleine Broekema
- Department of Human Genetics, Section Clinical Genetic, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Johanna M van Hagen
- Department of Human Genetics, Section Clinical Genetic, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Katheryn Grand
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Emily Magness
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Scott D McLean
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Division of Clinical Genetics, Christus Children's, San Antonio, TX
| | - Hülya Kayserili
- Department of Medical Genetics, Koç University School of Medicine, Istanbul, Turkey
| | - Umut Altunoglu
- Department of Medical Genetics, Koç University School of Medicine, Istanbul, Turkey
| | - Angie En Qi Chong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Shifeng Xue
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Médéric Jeanne
- Service de Génétique, FHU GenOMedS, CHRU de Tours, Tours, France; UMR1253, iBrain, Inserm, University of Tours, Tours, France
| | - Naif Almontashiri
- Center for Genetics and Inherited Diseases (CGID), Taibah University, Madinah, Saudi Arabia
| | - Wisam Habhab
- Department of Genetic Medicine, Faculty of Medicine, Princess Al-Jawhara Al-Brahim Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Laurence Faivre
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Centre Hospitalier Universitaire, Dijon, France; Genetics of Developmental Disorders, INSERM UMR1231, Université de Bourgogne, Dijon, France
| | - Eléonore Viora-Dupont
- Genetics of Developmental Disorders, INSERM UMR1231, Université de Bourgogne, Dijon, France; Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Centre Hospitalier Universitaire, Dijon, France
| | - Christophe Philippe
- Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Centre Hospitalier Universitaire, Dijon, France; Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon, Dijon, France
| | - Hana Safraou
- Centre de Référence Déficiences Intellectuelles de Causes Rares, FHU TRANSLAD, Centre Hospitalier Universitaire, Dijon, France; Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, CHU Dijon, Dijon, France
| | - Fanny Laffargue
- CHU Clermont Ferrand, Service de Génétique Clinique, Clermont Ferrand, France
| | - Luisa Mittendorf
- Department for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Ashwin Dalal
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Asodu Sandeep Sarma
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Boris Keren
- APHP, Sorbonne Université, Département de Génétique Médicale, GH Pitié Salpêtrière, Paris, France
| | - Bruno Reversade
- Laboratory of Human Genetics and Therapeutics, Genome Institute of Singapore (GIS), A∗STAR, Department of Physiology, Cardiovascular Disease, Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Medical Genetics, Koç University School of Medicine, Istanbul, Turkey; Laboratory of Human Genetics and Therapeutics Smart-Health Initiative, BESE, KAUST, Thuwal, Kingdom of Saudi Arabia
| | - Christèle Dubourg
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France; Génétique Moléculaire et Génomique, FHU GenOMedS, CHU de Rennes, Rennes, France
| | - Sylvie Odent
- Génétique Clinique, Centre de Référence Maladies Rares CLAD-Ouest, ERN-ITHACA, FHU GenOMedS, CHU de Rennes, Rennes, France; Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France
| | - Valérie Dupé
- Univ Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes)-UMR 6290, Rennes, France.
| |
Collapse
|
7
|
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
|
8
|
Schlissel G, Meziane M, Narducci D, Hansen AS, Li P. Diffusion barriers imposed by tissue topology shape morphogen gradients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592050. [PMID: 38746265 PMCID: PMC11092646 DOI: 10.1101/2024.05.01.592050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Animals use a small number of morphogens to pattern tissues, but it is unclear how evolution modulates morphogen signaling range to match tissues of varying sizes. Here, we used single molecule imaging in reconstituted morphogen gradients and in tissue explants to determine that Hedgehog diffused extra-cellularly as a monomer, and rapidly transitioned between membrane-confined and -unconfined states. Unexpectedly, the vertebrate-specific protein SCUBE1 expanded Hedgehog gradients by accelerating the transition rates between states without affecting the relative abundance of molecules in each state. This observation could not be explained under existing models of morphogen diffusion. Instead, we developed a topology-limited diffusion model in which cell-cell gaps create diffusion barriers, and morphogens can only overcome the barrier by passing through a membrane-unconfined state. Under this model, SCUBE1 promotes Hedgehog secretion and diffusion by allowing it to transiently overcome diffusion barriers. This multiscale understanding of morphogen gradient formation unified prior models and discovered novel knobs that nature can use to tune morphogen gradient sizes across tissues and organisms.
Collapse
|
9
|
Ehlers SF, Manikowski D, Steffes G, Ehring K, Gude F, Grobe K. A Residual N-Terminal Peptide Enhances Signaling of Depalmitoylated Hedgehog to the Patched Receptor. J Dev Biol 2024; 12:11. [PMID: 38651456 PMCID: PMC11036296 DOI: 10.3390/jdb12020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
During their biosynthesis, Sonic hedgehog (Shh) morphogens are covalently modified by cholesterol at the C-terminus and palmitate at the N-terminus. Although both lipids initially anchor Shh to the plasma membrane of producing cells, it later translocates to the extracellular compartment to direct developmental fates in cells expressing the Patched (Ptch) receptor. Possible release mechanisms for dually lipidated Hh/Shh into the extracellular compartment are currently under intense debate. In this paper, we describe the serum-dependent conversion of the dually lipidated cellular precursor into a soluble cholesteroylated variant (ShhC) during its release. Although ShhC is formed in a Dispatched- and Scube2-dependent manner, suggesting the physiological relevance of the protein, the depalmitoylation of ShhC during release is inconsistent with the previously postulated function of N-palmitate in Ptch receptor binding and signaling. Therefore, we analyzed the potency of ShhC to induce Ptch-controlled target cell transcription and differentiation in Hh-sensitive reporter cells and in the Drosophila eye. In both experimental systems, we found that ShhC was highly bioactive despite the absence of the N-palmitate. We also found that the artificial removal of N-terminal peptides longer than eight amino acids inactivated the depalmitoylated soluble proteins in vitro and in the developing Drosophila eye. These results demonstrate that N-depalmitoylated ShhC requires an N-peptide of a defined minimum length for its signaling function to Ptch.
Collapse
Affiliation(s)
- Sophia F. Ehlers
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Georg Steffes
- Institute for Neuro- and Behavioral Biology, Faculty of Biology, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany;
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| |
Collapse
|
10
|
Jiménez-Jiménez C, Grobe K, Guerrero I. Hedgehog on the Move: Glypican-Regulated Transport and Gradient Formation in Drosophila. Cells 2024; 13:418. [PMID: 38474382 PMCID: PMC10930589 DOI: 10.3390/cells13050418] [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: 01/30/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Glypicans (Glps) are a family of heparan sulphate proteoglycans that are attached to the outer plasma membrane leaflet of the producing cell by a glycosylphosphatidylinositol anchor. Glps are involved in the regulation of many signalling pathways, including those that regulate the activities of Wnts, Hedgehog (Hh), Fibroblast Growth Factors (FGFs), and Bone Morphogenetic Proteins (BMPs), among others. In the Hh-signalling pathway, Glps have been shown to be essential for ligand transport and the formation of Hh gradients over long distances, for the maintenance of Hh levels in the extracellular matrix, and for unimpaired ligand reception in distant recipient cells. Recently, two mechanistic models have been proposed to explain how Hh can form the signalling gradient and how Glps may contribute to it. In this review, we describe the structure, biochemistry, and metabolism of Glps and their interactions with different components of the Hh-signalling pathway that are important for the release, transport, and reception of Hh.
Collapse
Affiliation(s)
- Carlos Jiménez-Jiménez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Isabel Guerrero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| |
Collapse
|
11
|
Hall ET, Dillard ME, Cleverdon ER, Zhang Y, Daly CA, Ansari SS, Wakefield R, Stewart DP, Pruett-Miller SM, Lavado A, Carisey AF, Johnson A, Wang YD, Selner E, Tanes M, Ryu YS, Robinson CG, Steinberg J, Ogden SK. Cytoneme signaling provides essential contributions to mammalian tissue patterning. Cell 2024; 187:276-293.e23. [PMID: 38171360 PMCID: PMC10842732 DOI: 10.1016/j.cell.2023.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/06/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
During development, morphogens pattern tissues by instructing cell fate across long distances. Directly visualizing morphogen transport in situ has been inaccessible, so the molecular mechanisms ensuring successful morphogen delivery remain unclear. To tackle this longstanding problem, we developed a mouse model for compromised sonic hedgehog (SHH) morphogen delivery and discovered that endocytic recycling promotes SHH loading into signaling filopodia called cytonemes. We optimized methods to preserve in vivo cytonemes for advanced microscopy and show endogenous SHH localized to cytonemes in developing mouse neural tubes. Depletion of SHH from neural tube cytonemes alters neuronal cell fates and compromises neurodevelopment. Mutation of the filopodial motor myosin 10 (MYO10) reduces cytoneme length and density, which corrupts neuronal signaling activity of both SHH and WNT. Combined, these results demonstrate that cytoneme-based signal transport provides essential contributions to morphogen dispersion during mammalian tissue development and suggest MYO10 is a key regulator of cytoneme function.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Miriam E Dillard
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elizabeth R Cleverdon
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yan Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christina A Daly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shariq S Ansari
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Randall Wakefield
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alfonso Lavado
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alex F Carisey
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amanda Johnson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Emma Selner
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael Tanes
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Young Sang Ryu
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Camenzind G Robinson
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jeffrey Steinberg
- Center for In Vivo Imaging and Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
12
|
Varandas KC, Hodges BM, Lubeck L, Farinas A, Liang Y, Lu Y, Shaham S. Glia detect and mount a protective response to loss of dendrite substructure integrity in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567404. [PMID: 38014226 PMCID: PMC10680744 DOI: 10.1101/2023.11.16.567404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Neurons have elaborate structures that determine their connectivity and functions. Changes in neuronal structure accompany learning and memory formation and are hallmarks of neurological disease. Here we show that glia monitor dendrite structure and respond to dendrite perturbation. In C. elegans mutants with defective sensory-organ dendrite cilia, adjacent glia accumulate extracellular matrix-laden vesicles, secrete excess matrix around cilia, alter gene expression, and change their secreted protein repertoire. Inducible cilia disruption reveals that this response is acute. DGS-1, a 7-transmembrane domain neuronal protein, and FIG-1, a multifunctional thrombospondin-domain glial protein, are required for glial detection of cilia integrity, and exhibit mutually-dependent localization to and around cilia, respectively. While inhibiting glial secretion disrupts dendritic cilia properties, hyperactivating the glial response protects against dendrite damage. Our studies uncover a homeostatic protective dendrite-glia interaction and suggest that similar signaling occurs at other sensory structures and at synapses, which resemble sensory organs in architecture and molecules.
Collapse
|
13
|
Deshpande G, Ng C, Jourjine N, Chiew JW, Dasilva J, Schedl P. Hedgehog signaling guides migration of primordial germ cells to the Drosophila somatic gonad. Genetics 2023; 225:iyad165. [PMID: 37708366 PMCID: PMC10627259 DOI: 10.1093/genetics/iyad165] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
In addition to inducing nonautonomous specification of cell fate in both Drosophila and vertebrates, the Hedgehog pathway guides cell migration in a variety of different tissues. Although its role in axon guidance in the vertebrate nervous system is widely recognized, its role in guiding the migratory path of primordial germ cells (PGCs) from the outside surface of the Drosophila embryo through the midgut and mesoderm to the SGPs (somatic gonadal precursors) has been controversial. Here we present new experiments demonstrating (1) that Hh produced by mesodermal cells guides PGC migration, (2) that HMG CoenzymeA reductase (Hmgcr) potentiates guidance signals emanating from the SGPs, functioning upstream of hh and of 2 Hh pathway genes important for Hh-containing cytonemes, and (3) that factors required in Hh receiving cells in other contexts function in PGCs to help direct migration toward the SGPs. We also compare the data reported by 4 different laboratories that have studied the role of the Hh pathway in guiding PGC migration.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Chris Ng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Nicholas Jourjine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Joy Wan Chiew
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Juliana Dasilva
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
14
|
Tippetts TS, Sieber MH, Solmonson A. Beyond energy and growth: the role of metabolism in developmental signaling, cell behavior and diapause. Development 2023; 150:dev201610. [PMID: 37883062 PMCID: PMC10652041 DOI: 10.1242/dev.201610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Metabolism is crucial for development through supporting cell growth, energy production, establishing cell identity, developmental signaling and pattern formation. In many model systems, development occurs alongside metabolic transitions as cells differentiate and specialize in metabolism that supports new functions. Some cells exhibit metabolic flexibility to circumvent mutations or aberrant signaling, whereas other cell types require specific nutrients for developmental progress. Metabolic gradients and protein modifications enable pattern formation and cell communication. On an organism level, inadequate nutrients or stress can limit germ cell maturation, implantation and maturity through diapause, which slows metabolic activities until embryonic activation under improved environmental conditions.
Collapse
Affiliation(s)
- Trevor S. Tippetts
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew H. Sieber
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Laboratory of Developmental Metabolism and Placental Biology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Zhang Y, Beachy PA. Cellular and molecular mechanisms of Hedgehog signalling. Nat Rev Mol Cell Biol 2023; 24:668-687. [PMID: 36932157 DOI: 10.1038/s41580-023-00591-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 03/19/2023]
Abstract
The Hedgehog signalling pathway has crucial roles in embryonic tissue patterning, postembryonic tissue regeneration, and cancer, yet aspects of Hedgehog signal transmission and reception have until recently remained unclear. Biochemical and structural studies surprisingly reveal a central role for lipids in Hedgehog signalling. The signal - Hedgehog protein - is modified by cholesterol and palmitate during its biogenesis, thereby necessitating specialized proteins such as the transporter Dispatched and several lipid-binding carriers for cellular export and receptor engagement. Additional lipid transactions mediate response to the Hedgehog signal, including sterol activation of the transducer Smoothened. Access of sterols to Smoothened is regulated by the apparent sterol transporter and Hedgehog receptor Patched, whose activity is blocked by Hedgehog binding. Alongside these lipid-centric mechanisms and their relevance to pharmacological pathway modulation, we discuss emerging roles of Hedgehog pathway activity in stem cells or their cellular niches, with translational implications for regeneration and restoration of injured or diseased tissues.
Collapse
Affiliation(s)
- Yunxiao Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute and Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
16
|
Xu J, Iyyanar PPR, Lan Y, Jiang R. Sonic hedgehog signaling in craniofacial development. Differentiation 2023; 133:60-76. [PMID: 37481904 PMCID: PMC10529669 DOI: 10.1016/j.diff.2023.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Mutations in SHH and several other genes encoding components of the Hedgehog signaling pathway have been associated with holoprosencephaly syndromes, with craniofacial anomalies ranging in severity from cyclopia to facial cleft to midfacial and mandibular hypoplasia. Studies in animal models have revealed that SHH signaling plays crucial roles at multiple stages of craniofacial morphogenesis, from cranial neural crest cell survival to growth and patterning of the facial primordia to organogenesis of the palate, mandible, tongue, tooth, and taste bud formation and homeostasis. This article provides a summary of the major findings in studies of the roles of SHH signaling in craniofacial development, with emphasis on recent advances in the understanding of the molecular and cellular mechanisms regulating the SHH signaling pathway activity and those involving SHH signaling in the formation and patterning of craniofacial structures.
Collapse
Affiliation(s)
- Jingyue Xu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| | - Paul P R Iyyanar
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Yu Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
17
|
Ansell TB, Corey RA, Viti LV, Kinnebrew M, Rohatgi R, Siebold C, Sansom MSP. The energetics and ion coupling of cholesterol transport through Patched1. SCIENCE ADVANCES 2023; 9:eadh1609. [PMID: 37611095 PMCID: PMC10446486 DOI: 10.1126/sciadv.adh1609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023]
Abstract
Patched1 (PTCH1) is a tumor suppressor protein of the mammalian Hedgehog (HH) signaling pathway, implicated in embryogenesis and tissue homeostasis. PTCH1 inhibits the G protein-coupled receptor Smoothened (SMO) via a debated mechanism involving modulating ciliary cholesterol accessibility. Using extensive molecular dynamics simulations and free energy calculations to evaluate cholesterol transport through PTCH1, we find an energetic barrier of ~15 to 20 kilojoule per mole for cholesterol export. In silico data are coupled to in vivo biochemical assays of PTCH1 mutants to probe coupling between cation binding sites, transmembrane motions, and PTCH1 activity. Using complementary simulations of Dispatched1, we find that transition between "inward-open" and solvent "occluded" states is accompanied by Na+-induced pinching of intracellular helical segments. Thus, our findings illuminate the energetics and ion coupling stoichiometries of PTCH1 transport mechanisms, whereby one to three Na+ or two to three K+ couple to cholesterol export, and provide the first molecular description of transitions between distinct transport states.
Collapse
Affiliation(s)
- T Bertie Ansell
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Robin A Corey
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
- School of Physiology, Pharmacology and Neuroscience, Bristol University, Bristol BS8 1TD, UK
| | - Lucrezia Vittoria Viti
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Maia Kinnebrew
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
18
|
Bare Y, Matusek T, Vriz S, Deffieu MS, Thérond PP, Gaudin R. TMED10 mediates the loading of neosynthesised Sonic Hedgehog in COPII vesicles for efficient secretion and signalling. Cell Mol Life Sci 2023; 80:266. [PMID: 37624561 PMCID: PMC11072717 DOI: 10.1007/s00018-023-04918-1] [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: 04/18/2023] [Revised: 07/24/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
The morphogen Sonic Hedgehog (SHH) plays an important role in coordinating embryonic development. Short- and long-range SHH signalling occurs through a variety of membrane-associated and membrane-free forms. However, the molecular mechanisms that govern the early events of the trafficking of neosynthesised SHH in mammalian cells are still poorly understood. Here, we employed the retention using selective hooks (RUSH) system to show that newly-synthesised SHH is trafficked through the classical biosynthetic secretory pathway, using TMED10 as an endoplasmic reticulum (ER) cargo receptor for efficient ER-to-Golgi transport and Rab6 vesicles for Golgi-to-cell surface trafficking. TMED10 and SHH colocalized at ER exit sites (ERES), and TMED10 depletion significantly delays SHH loading onto ERES and subsequent exit leading to significant SHH release defects. Finally, we utilised the Drosophila wing imaginal disc model to demonstrate that the homologue of TMED10, Baiser (Bai), participates in Hedgehog (Hh) secretion and signalling in vivo. In conclusion, our work highlights the role of TMED10 in cargo-specific egress from the ER and sheds light on novel important partners of neosynthesised SHH secretion with potential impact on embryonic development.
Collapse
Affiliation(s)
- Yonis Bare
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS, 1919 Route de Mende, 34293, Montpellier, France
- Université de Montpellier, 34090, Montpellier, France
| | - Tamás Matusek
- Université Côte d'Azur, UMR7277 CNRS, Inserm 1091, Institut de Biologie de Valrose (iBV), Parc Valrose, Nice, France
| | - Sophie Vriz
- Laboratoire des Biomolécules (LBM), Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005, Paris, France
- Faculty of Science, Université de Paris, Paris, France
| | - Maika S Deffieu
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS, 1919 Route de Mende, 34293, Montpellier, France
- Université de Montpellier, 34090, Montpellier, France
| | - Pascal P Thérond
- Université Côte d'Azur, UMR7277 CNRS, Inserm 1091, Institut de Biologie de Valrose (iBV), Parc Valrose, Nice, France
| | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS, 1919 Route de Mende, 34293, Montpellier, France.
- Université de Montpellier, 34090, Montpellier, France.
| |
Collapse
|
19
|
Heimke M, Richter F, Heinze T, Kunke M, Wedel T, Böttner M, Egberts JH, Lucius R, Cossais F. Localization Pattern of Dispatched Homolog 2 (DISP2) in the Central and Enteric Nervous System. J Mol Neurosci 2023; 73:539-548. [PMID: 37369878 PMCID: PMC10517031 DOI: 10.1007/s12031-023-02129-8] [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: 03/07/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
Dispatched homolog (DISP) proteins have been implicated in the regulation of hedgehog signaling during embryologic development. Although DISP2 has recently been associated with neuronal development and control of cognitive functions, its localization pattern in the mammalian central and peripheral nervous system has not yet been investigated. In this study, the Disp2 expression profile was assessed in human tissues from publicly available transcriptomic datasets. The DISP2 localization pattern was further characterized in the human and rat central nervous system (CNS), as well as within the colonic enteric nervous system (ENS) using dual-label immunohistochemistry. Colocalization of DISP2 with neuronal and glial markers was additionally analyzed in murine primary ENS culture. At transcriptomic level, DISP2 expression was predominant in neuronal cell types of the CNS and ENS. DISP2 immunoreactivity was mainly located within PGP9.5-positive neurons rather than in S100-positive glial cells throughout the nervous system. Investigation of human and rat brain tissues, colonic specimens, and murine ENS primary cultures revealed that DISP2 was located in neuronal cell somata, as well as along neuronal processes both in the human and murine CNS and ENS. Our results indicate that DISP2 is prominently localized within neuronal cells of the CNS and ENS and support putative functions of DISP2 in these tissues.
Collapse
Affiliation(s)
- Marvin Heimke
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Florian Richter
- Department of General, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Tillmann Heinze
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Madlen Kunke
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Thilo Wedel
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - Martina Böttner
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | | | - Ralph Lucius
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany
| | - François Cossais
- Institute of Anatomy, Kiel University, Olshausenstrasse 40, 24098, Kiel, Germany.
| |
Collapse
|
20
|
Phénix J, Côté J, Dieme D, Recinto SJ, Oestereich F, Efrem S, Haddad S, Bouchard M, Munter LM. CETP inhibitor evacetrapib enters mouse brain tissue. Front Pharmacol 2023; 14:1171937. [PMID: 37533630 PMCID: PMC10390775 DOI: 10.3389/fphar.2023.1171937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
High levels of plasma cholesterol, especially high levels of low-density lipoprotein cholesterol (LDL-C), have been associated with an increased risk of Alzheimer's disease. The cholesteryl ester transfer protein (CETP) in plasma distributes cholesteryl esters between lipoproteins and increases LDL-C in plasma. Epidemiologically, decreased CETP activity has been associated with sustained cognitive performance during aging, longevity, and a lower risk of Alzheimer's disease. Thus, pharmacological CETP inhibitors could be repurposed for the treatment of Alzheimer's disease as they are safe and effective at lowering CETP activity and LDL-C. Although CETP is mostly expressed by the liver and secreted into the bloodstream, it is also expressed by astrocytes in the brain. Therefore, it is important to determine whether CETP inhibitors can enter the brain. Here, we describe the pharmacokinetic parameters of the CETP inhibitor evacetrapib in the plasma, liver, and brain tissues of CETP transgenic mice. We show that evacetrapib crosses the blood-brain barrier and is detectable in brain tissue 0.5 h after a 40 mg/kg i.v. injection in a non-linear function. We conclude that evacetrapib may prove to be a good candidate to treat CETP-mediated cholesterol dysregulation in Alzheimer's disease.
Collapse
Affiliation(s)
- Jasmine Phénix
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Cell Information Systems Group, Montreal, QC, Canada
| | - Jonathan Côté
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, QC, Canada
- Public Health Research Center (CReSP), Université de Montréal, Montreal, QC, Canada
| | - Denis Dieme
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, QC, Canada
- Public Health Research Center (CReSP), Université de Montréal, Montreal, QC, Canada
| | - Sherilyn J. Recinto
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Cell Information Systems Group, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Felix Oestereich
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Cell Information Systems Group, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Sasen Efrem
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Cell Information Systems Group, Montreal, QC, Canada
| | - Sami Haddad
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, QC, Canada
- Public Health Research Center (CReSP), Université de Montréal, Montreal, QC, Canada
| | - Michèle Bouchard
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, QC, Canada
- Public Health Research Center (CReSP), Université de Montréal, Montreal, QC, Canada
| | - Lisa Marie Munter
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Cell Information Systems Group, Montreal, QC, Canada
- Centre de Recherche en Biologie Structurale (CRBS), Montreal, QC, Canada
| |
Collapse
|
21
|
Abstract
Ligands of the Hedgehog (HH) pathway are paracrine signaling molecules that coordinate tissue development in metazoans. A remarkable feature of HH signaling is the repeated use of cholesterol in steps spanning ligand biogenesis, secretion, dispersal, and reception on target cells. A cholesterol molecule covalently attached to HH ligands is used as a molecular baton by transfer proteins to guide their secretion, spread, and reception. On target cells, a signaling circuit composed of a cholesterol transporter and sensor regulates transmission of HH signals across the plasma membrane to the cytoplasm. The repeated use of cholesterol in signaling supports the view that the HH pathway likely evolved by coopting ancient systems to regulate the abundance or organization of sterol-like lipids in membranes.
Collapse
Affiliation(s)
- Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom;
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, California, USA;
| |
Collapse
|
22
|
Lin WH, Cooper LM, Anastasiadis PZ. Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front Cell Dev Biol 2023; 11:1137013. [PMID: 37255594 PMCID: PMC10225604 DOI: 10.3389/fcell.2023.1137013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Cadherin-catenin complexes are integral components of the adherens junctions crucial for cell-cell adhesion and tissue homeostasis. Dysregulation of these complexes is linked to cancer development via alteration of cell-autonomous oncogenic signaling pathways and extrinsic tumor microenvironment. Advances in multiomics have uncovered key signaling events in multiple cancer types, creating a need for a better understanding of the crosstalk between cadherin-catenin complexes and oncogenic pathways. In this review, we focus on the biological functions of classical cadherins and associated catenins, describe how their dysregulation influences major cancer pathways, and discuss feedback regulation mechanisms between cadherin complexes and cellular signaling. We discuss evidence of cross regulation in the following contexts: Hippo-Yap/Taz and receptor tyrosine kinase signaling, key pathways involved in cell proliferation and growth; Wnt, Notch, and hedgehog signaling, key developmental pathways involved in human cancer; as well as TGFβ and the epithelial-to-mesenchymal transition program, an important process for cancer cell plasticity. Moreover, we briefly explore the role of cadherins and catenins in mechanotransduction and the immune tumor microenvironment.
Collapse
|
23
|
Ansell TB, Corey RA, Viti LV, Kinnebrew M, Rohatgi R, Siebold C, Sansom MSP. The Energetics and Ion Coupling of Cholesterol Transport Through Patched1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528445. [PMID: 36824746 PMCID: PMC9949057 DOI: 10.1101/2023.02.14.528445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Patched1 (PTCH1) is the principal tumour suppressor protein of the mammalian Hedgehog (HH) signalling pathway, implicated in embryogenesis and tissue homeostasis. PTCH1 inhibits the Class F G protein-coupled receptor Smoothened (SMO) via a debated mechanism involving modulating accessible cholesterol levels within ciliary membranes. Using extensive molecular dynamics (MD) simulations and free energy calculations to evaluate cholesterol transport through PTCH1, we find an energetic barrier of ~15-20 kJ mol -1 for cholesterol export. In simulations we identify cation binding sites within the PTCH1 transmembrane domain (TMD) which may provide the energetic impetus for cholesterol transport. In silico data are coupled to in vivo biochemical assays of PTCH1 mutants to probe coupling between transmembrane motions and PTCH1 activity. Using complementary simulations of Dispatched1 (DISP1) we find that transition between 'inward-open' and solvent 'occluded' states is accompanied by Na + induced pinching of intracellular helical segments. Thus, our findings illuminate the energetics and ion-coupling stoichiometries of PTCH1 transport mechanisms, whereby 1-3 Na + or 2-3 K + couple to cholesterol export, and provide the first molecular description of transitions between distinct transport states.
Collapse
Affiliation(s)
- T. Bertie Ansell
- Department of Biochemistry, South Parks Road, Oxford, OX1 3QU, UK
| | - Robin A. Corey
- Department of Biochemistry, South Parks Road, Oxford, OX1 3QU, UK
| | - Lucrezia Vittoria Viti
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Maia Kinnebrew
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | | |
Collapse
|
24
|
Manikowski D, Steffes G, Froese J, Exner S, Ehring K, Gude F, Di Iorio D, Wegner SV, Grobe K. Drosophila hedgehog signaling range and robustness depend on direct and sustained heparan sulfate interactions. Front Mol Biosci 2023; 10:1130064. [PMID: 36911531 PMCID: PMC9992881 DOI: 10.3389/fmolb.2023.1130064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Morphogens determine cellular differentiation in many developing tissues in a concentration dependent manner. As a central model for gradient formation during animal development, Hedgehog (Hh) morphogens spread away from their source to direct growth and pattern formation in the Drosophila wing disc. Although heparan sulfate (HS) expression in the disc is essential for this process, it is not known whether HS regulates Hh signaling and spread in a direct or in an indirect manner. To answer this question, we systematically screened two composite Hh binding areas for HS in vitro and expressed mutated proteins in the Drosophila wing disc. We found that selectively impaired HS binding of the second site reduced Hh signaling close to the source and caused striking wing mispatterning phenotypes more distant from the source. These observations suggest that HS constrains Hh to the wing disc epithelium in a direct manner, and that interfering with this constriction converts Hh into freely diffusing forms with altered signaling ranges and impaired gradient robustness.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Georg Steffes
- Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Sebastian Exner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Daniele Di Iorio
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Seraphine V Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
25
|
Wang J, Cui B, Li X, Zhao X, Huang T, Ding X. The emerging roles of Hedgehog signaling in tumor immune microenvironment. Front Oncol 2023; 13:1171418. [PMID: 37213270 PMCID: PMC10196179 DOI: 10.3389/fonc.2023.1171418] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
The Hedgehog (Hh) signaling pathway is pervasively involved in human malignancies, making it an effective target for cancer treatment for decades. In addition to its direct role in regulating cancer cell attributes, recent work indicates that it has an immunoregulatory effect on tumor microenvironments. An integrated understanding of these actions of Hh signaling pathway in tumor cells and tumor microenvironments will pave the way for novel tumor treatments and further advances in anti-tumor immunotherapy. In this review, we discuss the most recent research about Hh signaling pathway transduction, with a particular emphasis on its role in modulating tumor immune/stroma cell phenotype and function, such as macrophage polarity, T cell response, and fibroblast activation, as well as their mutual interactions between tumor cells and nonneoplastic cells. We also summarize the recent advances in the development of Hh pathway inhibitors and nanoparticle formulation for Hh pathway modulation. We suggest that targeting Hh signaling effects on both tumor cells and tumor immune microenvironments could be more synergistic for cancer treatment.
Collapse
Affiliation(s)
- Juan Wang
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Baiping Cui
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Xiaojie Li
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Xinyue Zhao
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Taomin Huang
- Department of Pharmacy, Eye & ENT Hospital, Fudan University, Shanghai, China
- *Correspondence: Taomin Huang, ; Xiaolei Ding,
| | - Xiaolei Ding
- Institute of Geriatrics, Affiliated Nantong Hospital of Shanghai University (The Sixth People’s Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
- *Correspondence: Taomin Huang, ; Xiaolei Ding,
| |
Collapse
|
26
|
Hedgehog-mediated gut-taste neuron axis controls sweet perception in Drosophila. Nat Commun 2022; 13:7810. [PMID: 36535958 PMCID: PMC9763350 DOI: 10.1038/s41467-022-35527-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Dietary composition affects food preference in animals. High sugar intake suppresses sweet sensation from insects to humans, but the molecular basis of this suppression is largely unknown. Here, we reveal that sugar intake in Drosophila induces the gut to express and secrete Hedgehog (Hh) into the circulation. We show that the midgut secreted Hh localize to taste sensilla and suppresses sweet sensation, perception, and preference. We further find that the midgut Hh inhibits Hh signalling in the sweet taste neurons. Our electrophysiology studies demonstrate that the midgut Hh signal also suppresses bitter taste and some odour responses, affecting overall food perception and preference. We further show that the level of sugar intake during a critical window early in life, sets the adult gut Hh expression and sugar perception. Our results together reveal a bottom-up feedback mechanism involving a "gut-taste neuron axis" that regulates food sensation and preference.
Collapse
|
27
|
Sandoval L, Labarca M, Retamal C, Sánchez P, Larraín J, González A. Sonic hedgehog is basolaterally sorted from the TGN and transcytosed to the apical domain involving Dispatched-1 at Rab11-ARE. Front Cell Dev Biol 2022; 10:833175. [PMID: 36568977 PMCID: PMC9768590 DOI: 10.3389/fcell.2022.833175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
Hedgehog proteins (Hhs) secretion from apical and/or basolateral domains occurs in different epithelial cells impacting development and tissue homeostasis. Palmitoylation and cholesteroylation attach Hhs to membranes, and Dispatched-1 (Disp-1) promotes their release. How these lipidated proteins are handled by the complex secretory and endocytic pathways of polarized epithelial cells remains unknown. We show that polarized Madin-Darby canine kidney cells address newly synthesized sonic hedgehog (Shh) from the TGN to the basolateral cell surface and then to the apical domain through a transcytosis pathway that includes Rab11-apical recycling endosomes (Rab11-ARE). Both palmitoylation and cholesteroylation contribute to this sorting behavior, otherwise Shh lacking these lipid modifications is secreted unpolarized. Disp-1 mediates first basolateral secretion from the TGN and then transcytosis from Rab11-ARE. At the steady state, Shh predominates apically and can be basolaterally transcytosed. This Shh trafficking provides several steps for regulation and variation in different epithelia, subordinating the apical to the basolateral secretion.
Collapse
Affiliation(s)
- Lisette Sandoval
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Mariana Labarca
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile,Centro Ciencia y Vida, Fundación Ciencia para la Vida, Santiago, Chile
| | - Claudio Retamal
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile,Centro Ciencia y Vida, Fundación Ciencia para la Vida, Santiago, Chile
| | - Paula Sánchez
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Larraín
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile,Centro Ciencia y Vida, Fundación Ciencia para la Vida, Santiago, Chile,Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile,*Correspondence: Alfonso González,
| |
Collapse
|
28
|
Jiang J. Hedgehog signaling mechanism and role in cancer. Semin Cancer Biol 2022; 85:107-122. [PMID: 33836254 PMCID: PMC8492792 DOI: 10.1016/j.semcancer.2021.04.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022]
Abstract
Cell-cell communication through evolutionarily conserved signaling pathways governs embryonic development and adult tissue homeostasis. Deregulation of these signaling pathways has been implicated in a wide range of human diseases including cancer. One such pathway is the Hedgehog (Hh) pathway, which was originally discovered in Drosophila and later found to play a fundamental role in human development and diseases. Abnormal Hh pathway activation is a major driver of basal cell carcinomas (BCC) and medulloblastoma. Hh exerts it biological influence through a largely conserved signal transduction pathway from the activation of the GPCR family transmembrane protein Smoothened (Smo) to the conversion of latent Zn-finger transcription factors Gli/Ci proteins from their repressor (GliR/CiR) to activator (GliA/CiA) forms. Studies from model organisms and human patients have provided deep insight into the Hh signal transduction mechanisms, revealed roles of Hh signaling in a wide range of human cancers, and suggested multiple strategies for targeting this pathway in cancer treatment.
Collapse
Affiliation(s)
- Jin Jiang
- Department of Molecular Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| |
Collapse
|
29
|
Bolatto C, Nieves S, Reyes A, Olivera-Bravo S, Cambiazo V. Patched-Related Is Required for Proper Development of Embryonic Drosophila Nervous System. Front Neurosci 2022; 16:920670. [PMID: 36081658 PMCID: PMC9446084 DOI: 10.3389/fnins.2022.920670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022] Open
Abstract
Patched-related (Ptr), classified primarily as a neuroectodermal gene, encodes a protein with predicted topology and domain organization closely related to those of Patched (Ptc), the canonical receptor of the Hedgehog (Hh) pathway. To investigate the physiological function of Ptr in the developing nervous system, Ptr null mutant embryos were immunolabeled and imaged under confocal microscopy. These embryos displayed severe alterations in the morphology of the primary axonal tracts, reduced number, and altered distribution of the Repo-positive glia as well as peripheral nervous system defects. Most of these alterations were recapitulated by downregulating Ptr expression, specifically in embryonic nerve cells. Because similar nervous system phenotypes have been observed in hh and ptc mutant embryos, we evaluated the Ptr participation in the Hh pathway by performing cell-based reporter assays. Clone-8 cells were transfected with Ptr-specific dsRNA or a Ptr DNA construct and assayed for changes in Hh-mediated induction of a luciferase reporter. The results obtained suggest that Ptr could act as a negative regulator of Hh signaling. Furthermore, co-immunoprecipitation assays from cell culture extracts premixed with a conditioned medium revealed a direct interaction between Ptr and Hh. Moreover, in vivo Ptr overexpression in the domain of the imaginal wing disc where Engrailed and Ptc coexist produced wing phenotypes at the A/P border. Thus, these results strongly suggest that Ptr plays a crucial role in nervous system development and appears to be a negative regulator of the Hh pathway.
Collapse
Affiliation(s)
- Carmen Bolatto
- Developmental Biology Laboratory, Histology and Embryology Department, Faculty of Medicine, Universidad de la República (UdelaR), Montevideo, Uruguay
- Cell and Molecular Neurobiology Laboratory, Computational and Integrative Neuroscience (NCIC) Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
- *Correspondence: Carmen Bolatto
| | - Sofía Nieves
- Developmental Biology Laboratory, Histology and Embryology Department, Faculty of Medicine, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Agustina Reyes
- Developmental Biology Laboratory, Histology and Embryology Department, Faculty of Medicine, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Silvia Olivera-Bravo
- Cell and Molecular Neurobiology Laboratory, Computational and Integrative Neuroscience (NCIC) Department, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Verónica Cambiazo
- Bioinformatic and Gene Expression Laboratory, Institute of Nutrition and Food Technology (INTA)-Universidad de Chile and Millennium Institute Center for Genome Regulation (CRG), Santiago, Chile
| |
Collapse
|
30
|
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: 2] [Impact Index Per Article: 0.7] [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
|
31
|
Abstract
Hedgehog (Hh) proteins constitute one family of a small number of secreted signaling proteins that together regulate multiple aspects of animal development, tissue homeostasis and regeneration. Originally uncovered through genetic analyses in Drosophila, their subsequent discovery in vertebrates has provided a paradigm for the role of morphogens in positional specification. Most strikingly, the Sonic hedgehog protein was shown to mediate the activity of two classic embryonic organizing centers in vertebrates and subsequent studies have implicated it and its paralogs in a myriad of processes. Moreover, dysfunction of the signaling pathway has been shown to underlie numerous human congenital abnormalities and diseases, especially certain types of cancer. This review focusses on the genetic studies that uncovered the key components of the Hh signaling system and the subsequent, biochemical, cell and structural biology analyses of their functions. These studies have revealed several novel processes and principles, shedding new light on the cellular and molecular mechanisms underlying cell-cell communication. Notable amongst these are the involvement of cholesterol both in modifying the Hh proteins and in activating its transduction pathway, the role of cytonemes, filipodia-like extensions, in conveying Hh signals between cells; and the central importance of the Primary Cilium as a cellular compartment within which the components of the signaling pathway are sequestered and interact.
Collapse
Affiliation(s)
- Philip William Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
32
|
Wang W, Shiraishi R, Kawauchi D. Sonic Hedgehog Signaling in Cerebellar Development and Cancer. Front Cell Dev Biol 2022; 10:864035. [PMID: 35573667 PMCID: PMC9100414 DOI: 10.3389/fcell.2022.864035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
The sonic hedgehog (SHH) pathway regulates the development of the central nervous system in vertebrates. Aberrant regulation of SHH signaling pathways often causes neurodevelopmental diseases and brain tumors. In the cerebellum, SHH secreted by Purkinje cells is a potent mitogen for granule cell progenitors, which are the most abundant cell type in the mature brain. While a reduction in SHH signaling induces cerebellar structural abnormalities, such as hypoplasia in various genetic disorders, the constitutive activation of SHH signaling often induces medulloblastoma (MB), one of the most common pediatric malignant brain tumors. Based on the existing literature on canonical and non-canonical SHH signaling pathways, emerging basic and clinical studies are exploring novel therapeutic approaches for MB by targeting SHH signaling at distinct molecular levels. In this review, we discuss the present consensus on SHH signaling mechanisms, their roles in cerebellar development and tumorigenesis, and the recent advances in clinical trials for MB.
Collapse
Affiliation(s)
- Wanchen Wang
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ryo Shiraishi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- Department of NCNP Brain Physiology and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Kawauchi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- *Correspondence: Daisuke Kawauchi,
| |
Collapse
|
33
|
Kaushal JB, Batra SK, Rachagani S. Hedgehog signaling and its molecular perspective with cholesterol: a comprehensive review. Cell Mol Life Sci 2022; 79:266. [PMID: 35486193 PMCID: PMC9990174 DOI: 10.1007/s00018-022-04233-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Hedgehog (Hh) signaling is evolutionarily conserved and plays an instructional role in embryonic morphogenesis, organogenesis in various animals, and the central nervous system organization. Multiple feedback mechanisms dynamically regulate this pathway in a spatiotemporal and context-dependent manner to confer differential patterns in cell fate determination. Hh signaling is complex due to canonical and non-canonical mechanisms coordinating cell-cell communication. In addition, studies have demonstrated a regulatory framework of Hh signaling and shown that cholesterol is vital for Hh ligand biogenesis, signal generation, and transduction from the cell surface to intracellular space. Studies have shown the importance of a specific cholesterol pool, termed accessible cholesterol, which serves as a second messenger, conveying signals between smoothened (Smo) and patched 1 (Ptch1) across the plasma and ciliary membranes. Remarkably, recent high-resolution structural and molecular studies shed new light on the interplay between Hh signaling and cholesterol in membrane biology. These studies elucidated novel mechanistic insight into the release and dispersal of cholesterol-anchored Hh and the basis of Hh recognition by Ptch1. Additionally, the putative model of Smo activation by cholesterol binding and/or modification and Ptch1 antagonization of Smo has been explicated. However, the coupling mechanism of Hh signaling and cholesterol offered a new regulatory principle in cell biology: how effector molecules of the Hh signal network react to and remodel cholesterol accessibility in the membrane and selectively activate Hh signaling proteins thereof. Recognizing the biological importance of cholesterol in Hh signaling activation and transduction opens the door for translational research to develop novel therapeutic strategies. This review looks in-depth at canonical and non-canonical Hh signaling and the distinct proposed model of cholesterol-mediated regulation of Hh signaling components, facilitating a more sophisticated understanding of the Hh signal network and cholesterol biology.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
34
|
Hanna CC, Kriegesmann J, Dowman LJ, Becker CFW, Payne RJ. Chemical Synthesis and Semisynthesis of Lipidated Proteins. Angew Chem Int Ed Engl 2022; 61:e202111266. [PMID: 34611966 PMCID: PMC9303669 DOI: 10.1002/anie.202111266] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Indexed: 11/24/2022]
Abstract
Lipidation is a ubiquitous modification of peptides and proteins that can occur either co- or post-translationally. An array of different lipid classes can adorn proteins and has been shown to influence a number of crucial biological activities, including the regulation of signaling, cell-cell adhesion events, and the anchoring of proteins to lipid rafts and phospholipid membranes. Whereas nature employs a range of enzymes to install lipid modifications onto proteins, the use of these for the chemoenzymatic generation of lipidated proteins is often inefficient or impractical. An alternative is to harness the power of modern synthetic and semisynthetic technologies to access lipid-modified proteins in a pure and homogeneously modified form. This Review aims to highlight significant advances in the development of lipidation and ligation chemistry and their implementation in the synthesis and semisynthesis of homogeneous lipidated proteins that have enabled the influence of these modifications on protein structure and function to be uncovered.
Collapse
Affiliation(s)
- Cameron C. Hanna
- School of ChemistryThe University of SydneySydneyNSW2006Australia
| | - Julia Kriegesmann
- Institute of Biological ChemistryFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Luke J. Dowman
- School of ChemistryThe University of SydneySydneyNSW2006Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of SydneySydneyNSW2006Australia
| | | | - Richard J. Payne
- School of ChemistryThe University of SydneySydneyNSW2006Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of SydneySydneyNSW2006Australia
| |
Collapse
|
35
|
Hanna CC, Kriegesmann J, Dowman LJ, Becker CFW, Payne RJ. Chemische Synthese und Semisynthese von lipidierten Proteinen. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202111266. [PMID: 38504765 PMCID: PMC10947004 DOI: 10.1002/ange.202111266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Indexed: 11/11/2022]
Abstract
AbstractLipidierung ist eine ubiquitäre Modifikation von Peptiden und Proteinen, die entweder co‐ oder posttranslational auftreten kann. Für die Vielzahl von Lipidklassen wurde gezeigt, dass diese viele entscheidende biologische Aktivitäten, z. B. die Regulierung der Signalweiterleitung, Zell‐Zell‐Adhäsion sowie die Anlagerung von Proteinen an Lipid‐Rafts und Phospholipidmembranen, beeinflussen. Während die Natur Enzyme nutzt, um Lipidmodifikationen in Proteine einzubringen, ist ihre Nutzung für die chemoenzymatische Herstellung von lipidierten Proteinen häufig ineffizient. Eine Alternative ist die Kombination moderner synthetischer und semisynthetischer Techniken, um lipidierte Proteine in reiner und homogen modifizierter Form zu erhalten. Dieser Aufsatz erörtert Fortschritte in der Entwicklung der Lipidierungs‐ und Ligationschemie und deren Anwendung in der Synthese und Semisynthese homogen lipidierter Proteine, die es ermöglichen, den Einfluss dieser Modifikationen auf die Proteinstruktur und ‐funktion zu untersuchen.
Collapse
Affiliation(s)
- Cameron C. Hanna
- School of ChemistryThe University of SydneySydneyNSW2006Australien
| | - Julia Kriegesmann
- Institut für Biologische ChemieFakultät für ChemieUniversität WienWienÖsterreich
| | - Luke J. Dowman
- School of ChemistryThe University of SydneySydneyNSW2006Australien
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of SydneySydneyNSW2006Australien
| | | | - Richard J. Payne
- School of ChemistryThe University of SydneySydneyNSW2006Australien
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of SydneySydneyNSW2006Australien
| |
Collapse
|
36
|
Garg C, khan H, Kaur A, Singh TG, Sharma VK, Singh SK. Therapeutic Implications of Sonic Hedgehog Pathway in Metabolic Disorders: Novel Target for Effective Treatment. Pharmacol Res 2022; 179:106194. [DOI: 10.1016/j.phrs.2022.106194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
|
37
|
Daly CA, Hall ET, Ogden SK. Regulatory mechanisms of cytoneme-based morphogen transport. Cell Mol Life Sci 2022; 79:119. [PMID: 35119540 PMCID: PMC8816744 DOI: 10.1007/s00018-022-04148-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/07/2023]
Abstract
During development and tissue homeostasis, cells must communicate with their neighbors to ensure coordinated responses to instructional cues. Cues such as morphogens and growth factors signal at both short and long ranges in temporal- and tissue-specific manners to guide cell fate determination, provide positional information, and to activate growth and survival responses. The precise mechanisms by which such signals traverse the extracellular environment to ensure reliable delivery to their intended cellular targets are not yet clear. One model for how this occurs suggests that specialized filopodia called cytonemes extend between signal-producing and -receiving cells to function as membrane-bound highways along which information flows. A growing body of evidence supports a crucial role for cytonemes in cell-to-cell communication. Despite this, the molecular mechanisms by which cytonemes are initiated, how they grow, and how they deliver specific signals are only starting to be revealed. Herein, we discuss recent advances toward improved understanding of cytoneme biology. We discuss similarities and differences between cytonemes and other types of cellular extensions, summarize what is known about how they originate, and discuss molecular mechanisms by which their activity may be controlled in development and tissue homeostasis. We conclude by highlighting important open questions regarding cytoneme biology, and comment on how a clear understanding of their function may provide opportunities for treating or preventing disease.
Collapse
Affiliation(s)
- Christina A Daly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, MS 1500, Memphis, TN, 38105, USA
| | - Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA.
| |
Collapse
|
38
|
Analysis of Dispatched Protein Processing and Sonic Hedgehog Ligand Release. Methods Mol Biol 2022; 2374:95-106. [PMID: 34562246 PMCID: PMC8864978 DOI: 10.1007/978-1-0716-1701-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The 12-pass transmembrane protein Dispatched (DISP) is essential for Sonic Hedgehog (SHH) release from ligand-producing cells and is indispensable for establishment of the SHH morphogen gradient during tissue patterning. Regulatory events controlling DISP release of SHH are not yet fully characterized. We recently demonstrated that DISP is cleaved by FURIN proprotein convertase at a conserved site in its first extracellular loop. Mutation of the cleavage site attenuates DISP-mediated SHH release, which indicates that Furin cleavage is a positive step toward DISP protein maturation. In this chapter, we present a ligand release/retention protocol that allows for the analysis of DISP cleavage, DISP-mediated release of SHH ligand from producing cells, and secretion-dependent signal induction in target cells.
Collapse
|
39
|
Hurbain I, Macé AS, Romao M, Prince E, Sengmanivong L, Ruel L, Basto R, Thérond PP, Raposo G, D'Angelo G. Microvilli-derived extracellular vesicles carry Hedgehog morphogenic signals for Drosophila wing imaginal disc development. Curr Biol 2021; 32:361-373.e6. [PMID: 34890558 DOI: 10.1016/j.cub.2021.11.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 08/12/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022]
Abstract
Morphogens are secreted molecules that regulate and coordinate major developmental processes, such as cell differentiation and tissue morphogenesis. Depending on the mechanisms of secretion and the nature of their carriers, morphogens act at short and long range. We investigated the paradigmatic long-range activity of Hedgehog (Hh), a well-known morphogen, and its contribution to the growth and patterning of the Drosophila wing imaginal disc. Extracellular vesicles (EVs) contribute to Hh long-range activity; however, the nature, the site, and the mechanisms underlying the biogenesis of these vesicular carriers remain unknown. Here, through the analysis of mutants and a series of Drosophila RNAi-depleted wing imaginal discs using fluorescence and live-imaging electron microscopy, including tomography and 3D reconstruction, we demonstrate that microvilli of the wing imaginal disc epithelium are the site of generation of small EVs that transport Hh across the tissue. Further, we show that the Prominin-like (PromL) protein is critical for microvilli integrity. Together with actin cytoskeleton and membrane phospholipids, PromL maintains microvilli architecture that is essential to promote its secretory function. Importantly, the distribution of Hh to microvilli and its release via these EVs contribute to the proper morphogenesis of the wing imaginal disc. Our results demonstrate that microvilli-derived EVs are carriers for Hh long-range signaling in vivo. By establishing that members of the Prominin protein family are key determinants of microvilli formation and integrity, our findings support the view that microvilli-derived EVs conveying Hh may provide a means for exchanging signaling cues of high significance in tissue development and cancer.
Collapse
Affiliation(s)
- Ilse Hurbain
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | - Anne-Sophie Macé
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | - Maryse Romao
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | - Elodie Prince
- Université Côte d'Azur, UMR7277 CNRS, Inserm U1091, Institute of Biology Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| | - Lucie Sengmanivong
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | - Laurent Ruel
- Université Côte d'Azur, UMR7277 CNRS, Inserm U1091, Institute of Biology Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| | - Renata Basto
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Pascal P Thérond
- Université Côte d'Azur, UMR7277 CNRS, Inserm U1091, Institute of Biology Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France
| | - Gisela D'Angelo
- Institut Curie, PSL Research University, CNRS, UMR144, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), 26, rue d'Ulm, 75248 Paris Cedex 05, France.
| |
Collapse
|
40
|
Li W, Wang L, Wierbowski BM, Lu M, Dong F, Liu W, Li S, Wang P, Salic A, Gong X. Structural insights into proteolytic activation of the human Dispatched1 transporter for Hedgehog morphogen release. Nat Commun 2021; 12:6966. [PMID: 34845226 PMCID: PMC8630017 DOI: 10.1038/s41467-021-27257-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022] Open
Abstract
The membrane protein Dispatched (Disp), which belongs to the RND family of small molecule transporters, is essential for Hedgehog (Hh) signaling, by catalyzing the extracellular release of palmitate- and cholesterol-modified Hh ligands from producing cells. Disp function requires Furin-mediated proteolytic cleavage of its extracellular domain, but how this activates Disp remains obscure. Here, we employ cryo-electron microscopy to determine atomic structures of human Disp1 (hDisp1), before and after cleavage, and in complex with lipid-modified Sonic hedgehog (Shh) ligand. These structures, together with biochemical data, reveal that proteolytic cleavage opens the extracellular domain of hDisp1, removing steric hindrance to Shh binding. Structure-guided functional experiments demonstrate the role of hDisp1-Shh interactions in ligand release. Our results clarify the mechanisms of hDisp1 activation and Shh morphogen release, and highlight how a unique proteolytic cleavage event enabled acquisition of a protein substrate by a member of a family of small molecule transporters.
Collapse
Affiliation(s)
- Wanqiu Li
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China ,grid.263817.90000 0004 1773 1790Present Address: Department of Pharmacology, School of Medicine, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Linlin Wang
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Bradley M. Wierbowski
- grid.38142.3c000000041936754XDepartment of Cell Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Mo Lu
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Feitong Dong
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Wenchen Liu
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Sisi Li
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China ,grid.508211.f0000 0004 6004 3854Present Address: Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, 518060 Shenzhen, Guangdong China
| | - Peiyi Wang
- grid.263817.90000 0004 1773 1790SUSTech Cryo-EM Facility Center, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Xin Gong
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
41
|
Dispatching plasma membrane cholesterol and Sonic Hedgehog dispatch: two sides of the same coin? Biochem Soc Trans 2021; 49:2455-2463. [PMID: 34515747 PMCID: PMC8589413 DOI: 10.1042/bst20210918] [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: 07/08/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Vertebrate and invertebrate Hedgehog (Hh) morphogens signal over short and long distances to direct cell fate decisions during development and to maintain tissue homeostasis after birth. One of the most important questions in Hh biology is how such Hh signaling to distant target cells is achieved, because all Hh proteins are secreted as dually lipidated proteins that firmly tether to the outer plasma membrane leaflet of their producing cells. There, Hhs multimerize into light microscopically visible storage platforms that recruit factors required for their regulated release. One such recruited release factor is the soluble glycoprotein Scube2 (Signal sequence, cubulin domain, epidermal-growth-factor-like protein 2), and maximal Scube2 function requires concomitant activity of the resistance-nodulation-division (RND) transporter Dispatched (Disp) at the plasma membrane of Hh-producing cells. Although recently published cryo-electron microscopy-derived structures suggest possible direct modes of Scube2/Disp-regulated Hh release, the mechanism of Disp-mediated Hh deployment is still not fully understood. In this review, we discuss suggested direct modes of Disp-dependent Hh deployment and relate them to the structural similarities between Disp and the related RND transporters Patched (Ptc) and Niemann-Pick type C protein 1. We then discuss open questions and perspectives that derive from these structural similarities, with particular focus on new findings that suggest shared small molecule transporter functions of Disp to deplete the plasma membrane of cholesterol and to modulate Hh release in an indirect manner.
Collapse
|
42
|
Wang Q, Asarnow DE, Ding K, Mann RK, Hatakeyama J, Zhang Y, Ma Y, Cheng Y, Beachy PA. Dispatched uses Na + flux to power release of lipid-modified Hedgehog. Nature 2021; 599:320-324. [PMID: 34707294 PMCID: PMC8785653 DOI: 10.1038/s41586-021-03996-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/06/2021] [Indexed: 01/02/2023]
Abstract
The Dispatched protein, which is related to the NPC1 and PTCH1 cholesterol transporters1,2 and to H+-driven transporters of the RND family3,4, enables tissue-patterning activity of the lipid-modified Hedgehog protein by releasing it from tightly -localized sites of embryonic expression5-10. Here we determine a cryo-electron microscopy structure of the mouse protein Dispatched homologue 1 (DISP1), revealing three Na+ ions coordinated within a channel that traverses its transmembrane domain. We find that the rate of Hedgehog export is dependent on the Na+ gradient across the plasma membrane. The transmembrane channel and Na+ binding are disrupted in DISP1-NNN, a variant with asparagine substitutions for three intramembrane aspartate residues that each coordinate and neutralize the charge of one of the three Na+ ions. DISP1-NNN and variants that disrupt single Na+ sites retain binding to, but are impaired in export of the lipid-modified Hedgehog protein to the SCUBE2 acceptor. Interaction of the amino-terminal signalling domain of the Sonic hedgehog protein (ShhN) with DISP1 occurs via an extensive buried surface area and contacts with an extended furin-cleaved DISP1 arm. Variability analysis reveals that ShhN binding is restricted to one extreme of a continuous series of DISP1 conformations. The bound and unbound DISP1 conformations display distinct Na+-site occupancies, which suggests a mechanism by which transmembrane Na+ flux may power extraction of the lipid-linked Hedgehog signal from the membrane. Na+-coordinating residues in DISP1 are conserved in PTCH1 and other metazoan RND family members, suggesting that Na+ flux powers their conformationally driven activities.
Collapse
Affiliation(s)
- Qianqian Wang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel E Asarnow
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Ke Ding
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Randall K Mann
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason Hatakeyama
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yunxiao Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Yong Ma
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Urology, and Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
43
|
Emerging roles of the Hedgehog signalling pathway in inflammatory bowel disease. Cell Death Discov 2021; 7:314. [PMID: 34702800 PMCID: PMC8548344 DOI: 10.1038/s41420-021-00679-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/15/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022] Open
Abstract
The Hedgehog (Hh) signalling pathway plays a critical role in the growth and patterning during embryonic development and maintenance of adult tissue homeostasis. Emerging data indicate that Hh signalling is implicated in the pathogenesis of inflammatory bowel disease (IBD). Current therapeutic treatments for IBD require optimisation, and novel effective drugs are warranted. Targeting the Hh signalling pathway may pave the way for successful IBD treatment. In this review, we introduce the molecular mechanisms underlying the Hh signalling pathway and its role in maintaining intestinal homeostasis. Then, we present interactions between the Hh signalling and other pathways involved in IBD and colitis-associated colorectal cancer (CAC), such as the Wnt and nuclear factor-kappa B (NF-κB) pathways. Furthermore, we summarise the latest research on Hh signalling associated with the occurrence and progression of IBD and CAC. Finally, we discuss the future directions for research on the role of Hh signalling in IBD pathogenesis and provide viewpoints on novel treatment options for IBD by targeting Hh signalling. An in-depth understanding of the complex role of Hh signalling in IBD pathogenesis will contribute to the development of new effective therapies for IBD patients.
Collapse
|
44
|
Manikowski D, Ehring K, Gude F, Jakobs P, Froese J, Grobe K. Hedgehog lipids: Promotors of alternative morphogen release and signaling?: Conflicting findings on lipidated Hedgehog transport and signaling can be explained by alternative regulated mechanisms to release the morphogen. Bioessays 2021; 43:e2100133. [PMID: 34611914 DOI: 10.1002/bies.202100133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022]
Abstract
Two posttranslational lipid modifications present on all Hedgehog (Hh) morphogens-an N-terminal palmitate and a C-terminal cholesterol-are established and essential regulators of Hh biofunction. Yet, for several decades, the question of exactly how both lipids contribute to Hh signaling remained obscure. Recently, cryogenic electron microscopy revealed different modes by which one or both lipids may contribute directly to Hh binding and signaling to its receptor Patched1 (Ptc). Some of these modes demand that the established release factor Dispatched1 (Disp) extracts dual-lipidated Hh from the cell surface, and that another known upstream signaling modulator called Scube2 chaperones the dual-lipidated morphogen to Ptc. By mechanistically and biochemically aligning this concept with established in vivo and recent in vitro findings, this reflection identifies remaining questions in lipidated Hh transport and evaluates additional mechanisms of Disp- and Scube2-regulated release of a second bioactive Hh fraction that has one or both lipids removed.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| |
Collapse
|
45
|
Luo Y, Wan G, Zhou X, Wang Q, Zhang Y, Bao J, Cong Y, Zhao Y, Li D. Architecture of Dispatched, a Transmembrane Protein Responsible for Hedgehog Release. Front Mol Biosci 2021; 8:701826. [PMID: 34557519 PMCID: PMC8453165 DOI: 10.3389/fmolb.2021.701826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/11/2021] [Indexed: 11/30/2022] Open
Abstract
The evolutionarily conserved Hedgehog (Hh) signaling pathway is crucial for programmed cell differentiation and proliferation. Dispatched (Disp) is a 12-transmembrane protein that plays a critical role in the Hedgehog (Hh) signaling pathway by releasing the dually lipidated ligand HhN from the membrane, a prerequisite step to the downstream signaling cascade. In this study, we focus on the Disp from water bear, a primitive animal known as the most indestructible on Earth. Using a zebrafish model, we show that the water bear homolog possesses the function of Disp. We have solved its structure to a 6.5-Å resolution using single-particle cryogenic electron microscopy. Consistent with the evolutional conservation of the pathway, the water bear Disp structure is overall similar to the previously reported structures of the fruit fly and human homologs. Although not revealing much detail at this resolution, the water bear Disp shows a different conformation compared to published structures, suggesting that they represent different functional snapshots.
Collapse
Affiliation(s)
- Yitian Luo
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guoyue Wan
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuan Zhou
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiuwen Wang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yunbin Zhang
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Bao
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yao Cong
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yun Zhao
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Dianfan Li
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
46
|
Ehring K, Manikowski D, Goretzko J, Froese J, Gude F, Jakobs P, Rescher U, Kirchhefer U, Grobe K. Conserved cholesterol-related activities of Dispatched 1 drive Sonic hedgehog shedding from the cell membrane. J Cell Sci 2021; 135:271842. [PMID: 34308968 PMCID: PMC8403983 DOI: 10.1242/jcs.258672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/08/2021] [Indexed: 01/04/2023] Open
Abstract
The Sonic hedgehog (Shh) pathway controls embryonic development and tissue homeostasis after birth. Long-standing questions about this pathway include how the dual-lipidated, firmly plasma membrane-associated Shh ligand is released from producing cells to signal to distant target cells and how the resistance-nodulation-division transporter Dispatched 1 (Disp, also known as Disp1) regulates this process. Here, we show that inactivation of Disp in Shh-expressing human cells impairs proteolytic Shh release from its lipidated terminal peptides, a process called ectodomain shedding. We also show that cholesterol export from Disp-deficient cells is reduced, that these cells contain increased cholesterol amounts in the plasma membrane, and that Shh shedding from Disp-deficient cells is restored by pharmacological membrane cholesterol extraction and by overexpression of transgenic Disp or the structurally related protein Patched 1 (Ptc, also known as Ptch1; a putative cholesterol transporter). These data suggest that Disp can regulate Shh function via controlled cell surface shedding and that membrane cholesterol-related molecular mechanisms shared by Disp and Ptc exercise such sheddase control.
Collapse
Affiliation(s)
- Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Jonas Goretzko
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Ursula Rescher
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of Münster, Domagkstrasse 12, D-48149 Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| |
Collapse
|
47
|
Hedgehog/GLI Signaling Pathway: Transduction, Regulation, and Implications for Disease. Cancers (Basel) 2021; 13:cancers13143410. [PMID: 34298625 PMCID: PMC8304605 DOI: 10.3390/cancers13143410] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The Hedgehog/GLI (Hh/GLI) pathway plays a major role during development and it is commonly dysregulated in many diseases, including cancer. This highly concerted series of ligands, receptors, cytoplasmic signaling molecules, transcription factors, and co-regulators is involved in regulating the biological functions controlled by this pathway. Activation of Hh/GLI in cancer is most often through a non-canonical method of activation, independent of ligand binding. This review is intended to summarize our current understanding of the Hh/GLI signaling, non-canonical mechanisms of pathway activation, its implication in disease, and the current therapeutic strategies targeting this cascade. Abstract The Hh/GLI signaling pathway was originally discovered in Drosophila as a major regulator of segment patterning in development. This pathway consists of a series of ligands (Shh, Ihh, and Dhh), transmembrane receptors (Ptch1 and Ptch2), transcription factors (GLI1–3), and signaling regulators (SMO, HHIP, SUFU, PKA, CK1, GSK3β, etc.) that work in concert to repress (Ptch1, Ptch2, SUFU, PKA, CK1, GSK3β) or activate (Shh, Ihh, Dhh, SMO, GLI1–3) the signaling cascade. Not long after the initial discovery, dysregulation of the Hh/GLI signaling pathway was implicated in human disease. Activation of this signaling pathway is observed in many types of cancer, including basal cell carcinoma, medulloblastoma, colorectal, prostate, pancreatic, and many more. Most often, the activation of the Hh/GLI pathway in cancer occurs through a ligand-independent mechanism. However, in benign disease, this activation is mostly ligand-dependent. The upstream signaling component of the receptor complex, SMO, is bypassed, and the GLI family of transcription factors can be activated regardless of ligand binding. Additional mechanisms of pathway activation exist whereby the entirety of the downstream signaling pathway is bypassed, and PTCH1 promotes cell cycle progression and prevents caspase-mediated apoptosis. Throughout this review, we summarize each component of the signaling cascade, non-canonical modes of pathway activation, and the implications in human disease, including cancer.
Collapse
|
48
|
Pizette S, Matusek T, Herpers B, Thérond PP, Rabouille C. Hherisomes, Hedgehog specialized recycling endosomes, are required for high level Hedgehog signaling and tissue growth. J Cell Sci 2021; 134:268340. [PMID: 34028543 DOI: 10.1242/jcs.258603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/07/2021] [Indexed: 01/03/2023] Open
Abstract
In metazoans, tissue growth and patterning is partly controlled by the Hedgehog (Hh) morphogen. Using immuno-electron microscopy on Drosophila wing imaginal discs, we identified a cellular structure, the Hherisomes, which contain the majority of intracellular Hh. Hherisomes are recycling tubular endosomes, and their formation is specifically boosted by overexpression of Hh. Expression of Rab11, a small GTPase involved in recycling endosomes, boosts the size of Hherisomes and their Hh concentration. Conversely, increased expression of the transporter Dispatched, a regulator of Hh secretion, leads to their clearance. We show that increasing Hh density in Hherisomes through Rab11 overexpression enhances both the level of Hh signaling and disc pouch growth, whereas Dispatched overexpression decreases high-level Hh signaling and growth. We propose that, upon secretion, a pool of Hh triggers low-level signaling, whereas a second pool of Hh is endocytosed and recycled through Hherisomes to stimulate high-level signaling and disc pouch growth. Altogether, our data indicate that Hherisomes are required to sustain physiological Hh activity necessary for patterning and tissue growth in the wing disc.
Collapse
Affiliation(s)
- Sandrine Pizette
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Inserm, Institute of Biology-Valrose (iBV), 06108 Nice Cedex 2, France
| | - Tamás Matusek
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Inserm, Institute of Biology-Valrose (iBV), 06108 Nice Cedex 2, France
| | - Bram Herpers
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Pascal P Thérond
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS), Inserm, Institute of Biology-Valrose (iBV), 06108 Nice Cedex 2, France
| | - Catherine Rabouille
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.,Hubrecht Institute/KNAW [Koninklijke Nederlandsee Akademie van Wetenschap (Dutch Royal Academy of Sciences)] and UMC Utrecht, 3584 CT Utrecht, The Netherlands.,Biological Sciences of Cells and Systems (BSBC) Department, UMC Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
49
|
Fitriasari S, Trainor PA. Diabetes, Oxidative Stress, and DNA Damage Modulate Cranial Neural Crest Cell Development and the Phenotype Variability of Craniofacial Disorders. Front Cell Dev Biol 2021; 9:644410. [PMID: 34095113 PMCID: PMC8174788 DOI: 10.3389/fcell.2021.644410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/21/2021] [Indexed: 12/11/2022] Open
Abstract
Craniofacial malformations are among the most common birth defects in humans and they often have significant detrimental functional, aesthetic, and social consequences. To date, more than 700 distinct craniofacial disorders have been described. However, the genetic, environmental, and developmental origins of most of these conditions remain to be determined. This gap in our knowledge is hampered in part by the tremendous phenotypic diversity evident in craniofacial syndromes but is also due to our limited understanding of the signals and mechanisms governing normal craniofacial development and variation. The principles of Mendelian inheritance have uncovered the etiology of relatively few complex craniofacial traits and consequently, the variability of craniofacial syndromes and phenotypes both within families and between families is often attributed to variable gene expression and incomplete penetrance. However, it is becoming increasingly apparent that phenotypic variation is often the result of combinatorial genetic and non-genetic factors. Major non-genetic factors include environmental effectors such as pregestational maternal diabetes, which is well-known to increase the risk of craniofacial birth defects. The hyperglycemia characteristic of diabetes causes oxidative stress which in turn can result in genotoxic stress, DNA damage, metabolic alterations, and subsequently perturbed embryogenesis. In this review we explore the importance of gene-environment associations involving diabetes, oxidative stress, and DNA damage during cranial neural crest cell development, which may underpin the phenotypic variability observed in specific craniofacial syndromes.
Collapse
Affiliation(s)
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
50
|
Yang S, Zhang Y, Yang C, Wu X, El Oud SM, Chen R, Cai X, Wu XS, Lan G, Zheng X. Competitive coordination of the dual roles of the Hedgehog co-receptor in homophilic adhesion and signal reception. eLife 2021; 10:65770. [PMID: 34003115 PMCID: PMC8131103 DOI: 10.7554/elife.65770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Hedgehog (Hh) signaling patterns embryonic tissues and contributes to homeostasis in adults. In Drosophila, Hh transport and signaling are thought to occur along a specialized class of actin-rich filopodia, termed cytonemes. Here, we report that Interference hedgehog (Ihog) not only forms a Hh receptor complex with Patched to mediate intracellular signaling, but Ihog also engages in trans-homophilic binding leading to cytoneme stabilization in a manner independent of its role as the Hh receptor. Both functions of Ihog (trans-homophilic binding for cytoneme stabilization and Hh binding for ligand sensing) involve a heparin-binding site on the first fibronectin repeat of the extracellular domain. Thus, the Ihog-Ihog interaction and the Hh-Ihog interaction cannot occur simultaneously for a single Ihog molecule. By combining experimental data and mathematical modeling, we determined that Hh-Ihog heterophilic interaction dominates and Hh can disrupt and displace Ihog molecules involved in trans-homophilic binding. Consequently, we proposed that the weaker Ihog-Ihog trans interaction promotes and stabilizes direct membrane contacts along cytonemes and that, as the cytoneme encounters secreted Hh ligands, the ligands trigger release of Ihog from trans Ihog-Ihog complex enabling transport or internalization of the Hh ligand-Ihog-Patched -receptor complex. Thus, the seemingly incompatible functions of Ihog in homophilic adhesion and ligand binding cooperate to assist Hh transport and reception along the cytonemes.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Ya Zhang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Chuxuan Yang
- Department of Physics, George Washington University, Washington, United States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Sarah Maria El Oud
- Department of Physics, George Washington University, Washington, United States
| | - Rongfang Chen
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xudong Cai
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xufeng S Wu
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Ganhui Lan
- Department of Physics, George Washington University, Washington, United States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| |
Collapse
|