1
|
Werder RB, Zhou X, Cho MH, Wilson AA. Breathing new life into the study of COPD with genes identified from genome-wide association studies. Eur Respir Rev 2024; 33:240019. [PMID: 38811034 PMCID: PMC11134200 DOI: 10.1183/16000617.0019-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 05/31/2024] Open
Abstract
COPD is a major cause of morbidity and mortality globally. While the significance of environmental exposures in disease pathogenesis is well established, the functional contribution of genetic factors has only in recent years drawn attention. Notably, many genes associated with COPD risk are also linked with lung function. Because reduced lung function precedes COPD onset, this association is consistent with the possibility that derangements leading to COPD could arise during lung development. In this review, we summarise the role of leading genes (HHIP, FAM13A, DSP, AGER and TGFB2) identified by genome-wide association studies in lung development and COPD. Because many COPD genome-wide association study genes are enriched in lung epithelial cells, we focus on the role of these genes in the lung epithelium in development, homeostasis and injury.
Collapse
Affiliation(s)
- Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, Australia
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
2
|
Wang H, Wang Y, Zhang H, Liang Z, Hu W, Qiu S, Li K, Zhang L, Dai H, Yang M, Yang G, Li L. Hedgehog interacting protein as a circulating biomarker in women with obesity: a cross-sectional study and intervention studies. Ann Med 2023; 55:2206162. [PMID: 37166403 PMCID: PMC10309011 DOI: 10.1080/07853890.2023.2206162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/18/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Although a study found a significant increase in serum hedgehog interacting protein (HHIP) concentrations in impaired fasting blood glucose, impaired glucose tolerance and newly diagnosed T2DM patients, the variation in circulating HHIP levels in obese individuals remains unknown. PATIENTS AND METHODS Gene Set Enrichment Analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used for differentially expressed genes and signal pathways. The study is comprised of a total of 452 young women, including 248 obese individuals and 204 controls. Circulating HHIP and Adipoq levels were determined with ELISA kits. Euglycemic-hyperinsulinemic clamps (EHC) and oral glucose tolerance test (OGTT) were conducted in every subject. 32 women were given metformin and 49 were given liraglutide treatment for 6 weeks. The study was registered with www.chictr.org.cn (ChiCTR2000032878 and ChiCTR1800019776). RESULTS Obesity was significantly associated with the cAMP signal pathway, and HHIP was a secreted protein related to cAMP signalling, as determined by KEGG analysis. In this population-based cohort study, we found that the level of circulating HHIP was significantly elevated in obese women, and positively correlated with body mass index and blood glucose, blood lipid, insulin, homeostasis model assessment of insulin resistance, dehydroepiandrostenedione sulphate, and luteinizing hormone, while negatively correlated with M-value and Adipoq. Insulin resistance (IR) and ove™rweight/obesity were associated with the higher HHIP concentration. OGTT and EHC tests revealed that the levels of circulating HHIP were regulated by blood glucose but to a less extent by insulin. After therapy with metformin and liraglutide, circulating HHIP levels were decreased, whereas Adipoq levels increased significantly. CONCLUSIONS Our findings support HHIP as a potential biomarker for predicting obesity and IR. In addition, drugs targeting HHIP may be a new strategy to treat obesity.
Collapse
Affiliation(s)
- Hao Wang
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yanping Wang
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongmin Zhang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Endocrinology, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Zerong Liang
- Department of Endocrinology, Chongqing Red Cross Hospital (People’s Hospital of Jiangbei District), Chongqing, China
| | - Wenjing Hu
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Sheng Qiu
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ke Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lili Zhang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Han Dai
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling Li
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Zhou X, Wang Y, Chen W, Zhang H, He Y, Dai H, Hu W, Li K, Zhang L, Chen C, Yang G, Li L. Circulating HHIP Levels in Women with Insulin Resistance and PCOS: Effects of Physical Activity, Cold Stimulation and Anti-Diabetic Drug Therapy. J Clin Med 2023; 12:888. [DOI: https:/doi.org/10.3390/jcm12030888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2024] Open
Abstract
Serum human hedgehog-interacting protein (HHIP) concentration is associated with diabetes. However, the relationship between HHIP and polycystic ovary syndrome (PCOS) or abnormal sex hormones remains unknown. This study was an observational cross-sectional study, with additional short-term intervention studies and follow-up studies. Bioinformatics analysis was performed to explore the association of PCOS with metabolic-related genes and signaling pathways. OGTT and EHC were performed on all participants. Lipid infusion, cold exposure, and 45-min treadmill test were performed on all healthy women. A total of 137 women with PCOS were treated with metformin, GLP-1RA, or TZDs for 24 weeks. Serum HHIP levels were higher in insulin resistance (IR) and PCOS women. Circulating HHIP levels were significantly correlated with adiponectin (Adipoq) levels, obesity, IR, and metabolic indicators. A correlation presented between HHIP and DHEA-S, FAI, SHBG, and FSH. Serum HHIP levels were significantly elevated by oral glucose challenge in healthy women, but not affected by EHC. Lipid infusion decreased serum HHIP levels, while cold exposure increased HHIP levels in healthy women. GLP-1RA and TZD treatment reduced serum HHIP levels in PCOS women, while metformin treatment did not affect HHIP levels. HHIP may be a useful biomarker and novel drug target for PCOS and IR individuals.
Collapse
Affiliation(s)
- Xin Zhou
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yanping Wang
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Wenyun Chen
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Hongmin Zhang
- Department of Endocrinology, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing 401121, China
| | - Yirui He
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Han Dai
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Wenjing Hu
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Ke Li
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Chen Chen
- Endocrinology and Metabolism, SBMS, The University of Queensland, Brisbane 4072, Australia
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Ling Li
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
4
|
Zhou X, Wang Y, Chen W, Zhang H, He Y, Dai H, Hu W, Li K, Zhang L, Chen C, Yang G, Li L. Circulating HHIP Levels in Women with Insulin Resistance and PCOS: Effects of Physical Activity, Cold Stimulation and Anti-Diabetic Drug Therapy. J Clin Med 2023; 12:888. [PMID: 36769536 PMCID: PMC9918013 DOI: 10.3390/jcm12030888] [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: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Serum human hedgehog-interacting protein (HHIP) concentration is associated with diabetes. However, the relationship between HHIP and polycystic ovary syndrome (PCOS) or abnormal sex hormones remains unknown. This study was an observational cross-sectional study, with additional short-term intervention studies and follow-up studies. Bioinformatics analysis was performed to explore the association of PCOS with metabolic-related genes and signaling pathways. OGTT and EHC were performed on all participants. Lipid infusion, cold exposure, and 45-min treadmill test were performed on all healthy women. A total of 137 women with PCOS were treated with metformin, GLP-1RA, or TZDs for 24 weeks. Serum HHIP levels were higher in insulin resistance (IR) and PCOS women. Circulating HHIP levels were significantly correlated with adiponectin (Adipoq) levels, obesity, IR, and metabolic indicators. A correlation presented between HHIP and DHEA-S, FAI, SHBG, and FSH. Serum HHIP levels were significantly elevated by oral glucose challenge in healthy women, but not affected by EHC. Lipid infusion decreased serum HHIP levels, while cold exposure increased HHIP levels in healthy women. GLP-1RA and TZD treatment reduced serum HHIP levels in PCOS women, while metformin treatment did not affect HHIP levels. HHIP may be a useful biomarker and novel drug target for PCOS and IR individuals.
Collapse
Affiliation(s)
- Xin Zhou
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yanping Wang
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Wenyun Chen
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Hongmin Zhang
- Department of Endocrinology, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing 401121, China
| | - Yirui He
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Han Dai
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Wenjing Hu
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| | - Ke Li
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Lili Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Chen Chen
- Endocrinology and Metabolism, SBMS, The University of Queensland, Brisbane 4072, Australia
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Ling Li
- The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
5
|
Rahi S, Mehan S. Understanding Abnormal SMO-SHH Signaling in Autism Spectrum Disorder: Potential Drug Target and Therapeutic Goals. Cell Mol Neurobiol 2022; 42:931-953. [PMID: 33206287 PMCID: PMC11441210 DOI: 10.1007/s10571-020-01010-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Autism is a multifactorial neurodevelopmental condition; it demonstrates some main characteristics, such as impaired social relationships and increased repetitive behavior. The initiation of autism spectrum disorder is mostly triggered during brain development by the deregulation of signaling pathways. Sonic hedgehog (SHH) signaling is one such mechanism that influences neurogenesis and neural processes during the development of the central nervous system. SMO-SHH signaling is also an important part of a broad variety of neurological processes, including neuronal cell differentiation, proliferation, and survival. Dysregulation of SMO-SHH signaling leads to many physiological changes that lead to neurological disorders such as ASD and contribute to cognitive decline. The aberrant downregulation of SMO-SHH signals contributes to the proteolytic cleavage of GLI (glioma-associated homolog) into GLI3 (repressor), which increases oxidative stress, neuronal excitotoxicity, neuroinflammation, and apoptosis by suppressing target gene expression. We outlined in this review that SMO-SHH deregulation plays a crucial role in the pathogenesis of autism and addresses the current status of SMO-SHH pathway modulators. Additionally, a greater understanding of the SHH signaling pathway is an effort to improve successful treatment for autism and other neurological disorders.
Collapse
Affiliation(s)
- Saloni Rahi
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
6
|
Griffiths SC, Schwab RA, El Omari K, Bishop B, Iverson EJ, Malinauskas T, Dubey R, Qian M, Covey DF, Gilbert RJC, Rohatgi R, Siebold C. Hedgehog-Interacting Protein is a multimodal antagonist of Hedgehog signalling. Nat Commun 2021; 12:7171. [PMID: 34887403 PMCID: PMC8660895 DOI: 10.1038/s41467-021-27475-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/19/2021] [Indexed: 01/20/2023] Open
Abstract
Hedgehog (HH) morphogen signalling, crucial for cell growth and tissue patterning in animals, is initiated by the binding of dually lipidated HH ligands to cell surface receptors. Hedgehog-Interacting Protein (HHIP), the only reported secreted inhibitor of Sonic Hedgehog (SHH) signalling, binds directly to SHH with high nanomolar affinity, sequestering SHH. Here, we report the structure of the HHIP N-terminal domain (HHIP-N) in complex with a glycosaminoglycan (GAG). HHIP-N displays a unique bipartite fold with a GAG-binding domain alongside a Cysteine Rich Domain (CRD). We show that HHIP-N is required to convey full HHIP inhibitory function, likely by interacting with the cholesterol moiety covalently linked to HH ligands, thereby preventing this SHH-attached cholesterol from binding to the HH receptor Patched (PTCH1). We also present the structure of the HHIP C-terminal domain in complex with the GAG heparin. Heparin can bind to both HHIP-N and HHIP-C, thereby inducing clustering at the cell surface and generating a high-avidity platform for SHH sequestration and inhibition. Our data suggest a multimodal mechanism, in which HHIP can bind two specific sites on the SHH morphogen, alongside multiple GAG interactions, to inhibit SHH signalling.
Collapse
Affiliation(s)
- Samuel C Griffiths
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Evotec (UK) Ltd., Milton Park, Abingdon, UK
| | - Rebekka A Schwab
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kamel El Omari
- Science Division, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellen J Iverson
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ramin Dubey
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MI, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MI, USA
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - 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, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Association of Sonic Hedgehog with the extracellular matrix requires its zinc-coordination center. BMC Mol Cell Biol 2021; 22:22. [PMID: 33863273 PMCID: PMC8052667 DOI: 10.1186/s12860-021-00359-5] [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: 11/18/2020] [Accepted: 03/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Sonic Hedgehog (Shh) has a catalytic cleft characteristic for zinc metallopeptidases and has significant sequence similarities with some bacterial peptidoglycan metallopeptidases defining a subgroup within the M15A family that, besides having the characteristic zinc coordination motif, can bind two calcium ions. Extracellular matrix (ECM) components in animals include heparan-sulfate proteoglycans, which are analogs of bacterial peptidoglycan and are involved in the extracellular distribution of Shh. Results We found that the zinc-coordination center of Shh is required for its association to the ECM as well as for non-cell autonomous signaling. Association with the ECM requires the presence of at least 0.1 μM zinc and is prevented by mutations affecting critical conserved catalytical residues. Consistent with the presence of a conserved calcium binding domain, we find that extracellular calcium inhibits ECM association of Shh. Conclusions Our results indicate that the putative intrinsic peptidase activity of Shh is required for non-cell autonomous signaling, possibly by enzymatically altering ECM characteristics. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00359-5.
Collapse
|
8
|
Tirou L, Russo M, Faure H, Pellegrino G, Demongin C, Daynac M, Sharif A, Amosse J, Le Lay S, Denis R, Luquet S, Taouis M, Benomar Y, Ruat M. Sonic Hedgehog receptor Patched deficiency in astrocytes enhances glucose metabolism in mice. Mol Metab 2021; 47:101172. [PMID: 33513436 PMCID: PMC7893488 DOI: 10.1016/j.molmet.2021.101172] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 01/06/2023] Open
Abstract
Objective Astrocytes are glial cells proposed as the main Sonic hedgehog (Shh)-responsive cells in the adult brain. Their roles in mediating Shh functions are still poorly understood. In the hypothalamus, astrocytes support neuronal circuits implicated in the regulation of energy metabolism. In this study, we investigated the impact of genetic activation of Shh signaling on hypothalamic astrocytes and characterized its effects on energy metabolism. Methods We analyzed the distribution of gene transcripts of the Shh pathway (Ptc, Gli1, Gli2, and Gli3) in astrocytes using single molecule fluorescence in situ hybridization combined with immunohistofluorescence of Shh peptides by Western blotting in the adult mouse hypothalamus. Based on the metabolic phenotype, we characterized Glast-CreERT2-YFP-Ptc−/− (YFP-Ptc−/−) mice and their controls over time and under a high-fat diet (HFD) to investigate the potential effects of conditional astrocytic deletion of the Shh receptor Patched (Ptc) on metabolic efficiency, insulin sensitivity, and systemic glucose metabolism. Molecular and biochemical assays were used to analyze the alteration of key pathways modulating energy metabolism, insulin sensitivity, glucose uptake, and inflammation. Primary astrocyte cultures were used to evaluate a potential role of Shh signaling in astrocytic glucose uptake. Results Shh peptides were the highest in the hypothalamic extracts of adult mice and a large population of hypothalamic astrocytes expressed Ptc and Gli1-3 mRNAs. Characterization of Shh signaling after conditional Ptc deletion in the YFP-Ptc−/− mice revealed heterogeneity in hypothalamic astrocyte populations. Interestingly, activation of Shh signaling in Glast+ astrocytes enhanced insulin responsiveness as evidenced by glucose and insulin tolerance tests. This effect was maintained over time and associated with lower blood insulin levels and also observed under a HFD. The YFP-Ptc−/− mice exhibited a lean phenotype with the absence of body weight gain and a marked reduction of white and brown adipose tissues accompanied by increased whole-body fatty acid oxidation. In contrast, food intake, locomotor activity, and body temperature were not altered. At the cellular level, Ptc deletion did not affect glucose uptake in primary astrocyte cultures. In the hypothalamus, activation of the astrocytic Shh pathway was associated with the upregulation of transcripts coding for the insulin receptor and liver kinase B1 (LKB1) after 4 weeks and the glucose transporter GLUT-4 after 32 weeks. Conclusions Here, we define hypothalamic Shh action on astrocytes as a novel master regulator of energy metabolism. In the hypothalamus, astrocytic Shh signaling could be critically involved in preventing both aging- and obesity-related metabolic disorders. Astrocytes exhibit differences in regulating the hedgehog signaling pathway. Deletion of Ptc in Glast+ cells prevents body weight gain and insulin resistance. Deletion of Ptc in Glast+ cells increases β oxidation. Central hedgehog signaling participates in the regulation of whole-body metabolism.
Collapse
Affiliation(s)
- Linda Tirou
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mariagiovanna Russo
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Helene Faure
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Giuliana Pellegrino
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Clement Demongin
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mathieu Daynac
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog (JPARC) - Lille Neurosciences & Cognition, F-59000, Lille, France
| | - Jeremy Amosse
- Univ. Angers SFR ICAT, F-49100, Angers, France; IRSET Laboratory, Inserm, UMR, 1085, Rennes, France
| | - Soazig Le Lay
- Univ. Angers SFR ICAT, F-49100, Angers, France; Univ. Nantes, CNRS, Inserm, Thorax Institut, F-44000, Nantes, France
| | - Raphaël Denis
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Serge Luquet
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Mohammed Taouis
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Yacir Benomar
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Martial Ruat
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France.
| |
Collapse
|
9
|
Hedgehog Interacting Protein (Hhip) Regulates Insulin Secretion in Mice Fed High Fat Diets. Sci Rep 2019; 9:11183. [PMID: 31371780 PMCID: PMC6673691 DOI: 10.1038/s41598-019-47633-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
Hedgehog interacting protein (Hhip) is essential for islet formation and beta-cell proliferation during pancreatic development; abnormally elevated Hhip expression has been linked to human pancreatitis. Here, we investigate the role of Hhip in modulating insulin secretion in adult Hhip mice (Hhip +/− vs. Hhip+/+) fed high fat diets (HFD). Both sexes of HFD-Hhip +/+ mice developed impaired glucose intolerance, that was only ameliorated in male HFD-Hhip +/− mice that had high levels of circulating plasma insulin, but not in female HFD-Hhip +/− mice. HFD stimulated Hhip gene expression, mainly in beta cells. Male HFD-Hhip +/+ mice had more large islets in which insulin content was reduced; islet architecture was disordered; and markers of oxidative stress (8-OHdG and Nox 2) were increased. In contrast, male HFD-Hhip +/− mice had more small islets with increased beta cell proliferation, enhanced GSIS, less oxidative stress and preserved islet integrity. In vitro, recombinant Hhip increased Nox2 and NADPH activity and decreased insulin-positive beta cells. siRNA-Hhip increased GSIS and abolished the stimulation of sodium palmitate (PA)-BSA on Nox2 gene expression. We conclude that pancreatic Hhip gene inhibits insulin secretion by altering islet integrity and promoting Nox2 gene expression in beta cells in response to HDF-mediated beta cell dysfunction, a novel finding.
Collapse
|
10
|
Wei H, Li J, Shi S, Zhang L, Xiang A, Shi X, Yang G, Chu G. Hhip inhibits proliferation and promotes differentiation of adipocytes through suppressing hedgehog signaling pathway. Biochem Biophys Res Commun 2019; 514:148-156. [PMID: 31027733 DOI: 10.1016/j.bbrc.2019.04.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/06/2019] [Indexed: 12/19/2022]
Abstract
Adipogenesis, which directly control body fat mass, plays a crucial role in lipid metabolism and obesity-related diseases. Hedgehog interacting protein (Hhip) belongs to Hedgehog (Hh) signaling pathway. The Hh signaling pathway was already linked with adipogenesis in previous reports, however, the physiological functions of Hhip on lipid deposition are still poorly understood. In this study, the level of Hhip was down-regulated during the development of porcine adipose tissues. Recombinant Hedgehog interacting protein (rHhip) could down-regulate cell cycle related genes and cell numbers in S phage to inhibit cell proliferation. Moreover, rHhip could increase adipocytes differentiation by targeting canonical Hh signaling, indicated by the increase of lipid accumulation and up-regulation of Glut4 and PPARγ expression. Collectively, these findings illustrated the essential role of Hhip in the proliferation and differentiation of adipocytes, and provided a potential novel target for preventing obesity.
Collapse
Affiliation(s)
- Haiyan Wei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jingjing Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shengjie Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lutong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Aoqi Xiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xin'e Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Guiyan Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
11
|
Yang Z, He LJ, Sun SR. Role of Endothelial Cells in Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:145-163. [PMID: 31399965 DOI: 10.1007/978-981-13-8871-2_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Renal fibrosis has been regarded as the common pathway of end-stage renal failure. Understanding the fundamental mechanism that leads to renal fibrosis is essential for developing better therapeutic options for chronic kidney diseases. So far, the main abstractions are on the injury of tubular epithelial cells, activation of interstitial cells, expression of chemotactic factor and adhesion molecule, infiltration of inflammatory cells and homeostasis of ECM. However, emerging studies revealed that endothelial cells (ECs) might happen to endothelial-to-mesenchymal transition (EndMT) dependent and/or independent endothelial dysfunction, which were supposed to accelerate renal fibrosis and are identified as new mechanisms for the proliferation of myofibroblasts as well. In this chapter, we are about to interpret the role of ECs in renal fibrosis and analyze the related molecules and pathways of both EndMT and EndMT independent endothelial dysfunction.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Nephrology, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Li-Jie He
- Department of Nephrology, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Shi-Ren Sun
- Department of Nephrology, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
12
|
Hedgehog Interacting Protein Promotes Fibrosis and Apoptosis in Glomerular Endothelial Cells in Murine Diabetes. Sci Rep 2018; 8:5958. [PMID: 29654303 PMCID: PMC5899163 DOI: 10.1038/s41598-018-24220-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
We investigated whether renal hedgehog interacting protein (Hhip) expression contributes to the progression of diabetic nephropathy (DN) and studied its related mechanism(s) in vivo and in vitro. Here, we show that Hhip expression is highly elevated in glomerular endothelial cells of adult type 1 diabetic (T1D) Akita and T2D db/db mouse kidneys as compared to non-diabetic control littermates. Hyperglycemia enhances reactive oxygen species (ROS) generation via NADPH oxidase 4 (Nox4) activation and stimulates renal Hhip gene expression, and that elevated renal Hhip gene expression subsequently activates the TGFβ1- Smad2/3 cascade and promotes endothelial to mesenchymal transition associated with endothelial cell fibrosis/apoptosis in vivo and in vitro. Furthermore, kidneys of low-dose streptozotocin-induced diabetic heterozygous Hhip deficient (Hhip+/−) mice displayed a normal albumin/creatinine ratio with fewer features of DN (glomerulosclerosis/fibrosis and podocyte apoptosis/loss) and less evidence of renal compensation (glomerular hypertrophy and hyperfiltration) as compared to diabetic wild type controls (Hhip+/+). Thus, our studies demonstrated that renal Hhip expression is associated with nephropathy development in diabetes and that hyperglycemia-induced renal Hhip expression may mediate glomerular endothelial fibrosis and apoptosis in diabetes, a novel finding.
Collapse
|
13
|
Liao MC, Zhao XP, Chang SY, Lo CS, Chenier I, Takano T, Ingelfinger JR, Zhang SL. AT 2 R deficiency mediated podocyte loss via activation of ectopic hedgehog interacting protein (Hhip) gene expression. J Pathol 2017; 243:279-293. [PMID: 28722118 DOI: 10.1002/path.4946] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/26/2017] [Accepted: 07/08/2017] [Indexed: 01/10/2023]
Abstract
Angiotensin II type 2 receptor (AT2 R) deficiency in AT2 R knockout (KO) mice has been linked to congenital abnormalities of the kidney and urinary tract; however, the mechanisms by which this occurs are poorly understood. In this study, we examined whether AT2 R deficiency impaired glomerulogenesis and mediated podocyte loss/dysfunction in vivo and in vitro. Nephrin-cyan fluorescent protein (CFP)-transgenic (Tg) and Nephrin/AT2 RKO mice were used to assess glomerulogenesis, while wild-type and AT2 RKO mice were used to evaluate maturation of podocyte morphology/function. Immortalized mouse podocytes (mPODs) were employed for in vitro studies. AT2 R deficiency resulted in diminished glomerulogenesis in E15 embryos, but had no impact on actual nephron number in neonates. Pups lacking AT2 R displayed features of renal dysplasia with lower glomerular tuft volume and podocyte numbers. In vivo and in vitro studies demonstrated that loss of AT2 R was associated with elevated NADPH oxidase 4 levels, which in turn stimulated ectopic hedgehog interacting protein (Hhip) gene expression in podocytes. Consequently, ectopic Hhip expression activation either triggers caspase-3 and p53-related apoptotic processes resulting in podocyte loss, or activates TGFβ1-Smad2/3 cascades and α-SMA expression to transform differentiated podocytes to undifferentiated podocyte-derived fibrotic cells. We analyzed HHIP expression in the kidney disease database (Nephroseq) and then validated this using HHIP immunohistochemistry staining of human kidney biopsies (controls versus focal segmental glomerulosclerosis). In conclusion, loss of AT2 R is associated with podocyte loss/dysfunction and is mediated, at least in part, via augmented ectopic Hhip expression in podocytes. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Min-Chun Liao
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| | - Xin-Ping Zhao
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| | - Shiao-Ying Chang
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| | - Chao-Sheng Lo
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| | - Isabelle Chenier
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| | - Tomoko Takano
- McGill University Health Centre, Montréal, Québec, Canada
| | - Julie R Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shao-Ling Zhang
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, Montréal, Québec, Canada
| |
Collapse
|
14
|
Patel SS, Tomar S, Sharma D, Mahindroo N, Udayabanu M. Targeting sonic hedgehog signaling in neurological disorders. Neurosci Biobehav Rev 2017; 74:76-97. [PMID: 28088536 DOI: 10.1016/j.neubiorev.2017.01.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/29/2016] [Accepted: 01/07/2017] [Indexed: 12/13/2022]
Abstract
Sonic hedgehog (Shh) signaling influences neurogenesis and neural patterning during the development of central nervous system. Dysregulation of Shh signaling in brain leads to neurological disorders like autism spectrum disorder, depression, dementia, stroke, Parkinson's diseases, Huntington's disease, locomotor deficit, epilepsy, demyelinating disease, neuropathies as well as brain tumors. The synthesis, processing and transport of Shh ligand as well as the localization of its receptors and signal transduction in the central nervous system has been carefully reviewed. Further, we summarize the regulation of small molecule modulators of Shh pathway with potential in neurological disorders. In conclusion, further studies are warranted to demonstrate the potential of positive and negative regulators of the Shh pathway in neurological disorders.
Collapse
Affiliation(s)
- Sita Sharan Patel
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India
| | - Sunil Tomar
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Diksha Sharma
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Neeraj Mahindroo
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan 173212, Himachal Pradesh, India
| | - Malairaman Udayabanu
- Department of Pharmacy, Jaypee University of Information Technology, Waknaghat 173234, Himachal Pradesh, India.
| |
Collapse
|
15
|
Holtz AM, Griffiths SC, Davis SJ, Bishop B, Siebold C, Allen BL. Secreted HHIP1 interacts with heparan sulfate and regulates Hedgehog ligand localization and function. J Cell Biol 2015; 209:739-57. [PMID: 26056142 PMCID: PMC4460154 DOI: 10.1083/jcb.201411024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/24/2015] [Indexed: 01/21/2023] Open
Abstract
Vertebrate Hedgehog (HH) signaling is controlled by several ligand-binding antagonists including Patched-1 (PTCH1), PTCH2, and HH-interacting protein 1 (HHIP1), whose collective action is essential for proper HH pathway activity. However, the molecular mechanisms used by these inhibitors remain poorly understood. In this paper, we investigated the mechanisms underlying HHIP1 antagonism of HH signaling. Strikingly, we found evidence that HHIP1 non-cell-autonomously inhibits HH-dependent neural progenitor patterning and proliferation. Furthermore, this non-cell-autonomous antagonism of HH signaling results from the secretion of HHIP1 that is modulated by cell type-specific interactions with heparan sulfate (HS). These interactions are mediated by an HS-binding motif in the cysteine-rich domain of HHIP1 that is required for its localization to the neuroepithelial basement membrane (BM) to effectively antagonize HH pathway function. Our data also suggest that endogenous, secreted HHIP1 localization to HS-containing BMs regulates HH ligand distribution. Overall, the secreted activity of HHIP1 represents a novel mechanism to regulate HH ligand localization and function during embryogenesis.
Collapse
Affiliation(s)
- Alexander M Holtz
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109 Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109 Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109
| | - Samuel C Griffiths
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England, UK
| | - Samantha J Davis
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England, UK
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
16
|
Hoch L, Faure H, Roudaut H, Schoenfelder A, Mann A, Girard N, Bihannic L, Ayrault O, Petricci E, Taddei M, Rognan D, Ruat M. MRT-92 inhibits Hedgehog signaling by blocking overlapping binding sites in the transmembrane domain of the Smoothened receptor. FASEB J 2015; 29:1817-29. [PMID: 25636740 DOI: 10.1096/fj.14-267849] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 12/18/2014] [Indexed: 12/28/2022]
Abstract
The Smoothened (Smo) receptor, a member of class F G protein-coupled receptors, is the main transducer of the Hedgehog (Hh) signaling pathway implicated in a wide range of developmental and adult processes. Smo is the target of anticancer drugs that bind to a long and narrow cavity in the 7-transmembrane (7TM) domain. X-ray structures of human Smo (hSmo) bound to several ligands have revealed 2 types of 7TM-directed antagonists: those binding mostly to extracellular loops (site 1, e.g., LY2940680) and those penetrating deeply in the 7TM cavity (site 2, e.g., SANT-1). Here we report the development of the acylguanidine MRT-92, which displays subnanomolar antagonist activity against Smo in various Hh cell-based assays. MRT-92 inhibits rodent cerebellar granule cell proliferation induced by Hh pathway activation through pharmacologic (half maximal inhibitory concentration [IC50] = 0.4 nM) or genetic manipulation. Using [(3)H]MRT-92 (Kd = 0.3 nM for hSmo), we created a comprehensive framework for the interaction of small molecule modulators with hSmo and for understanding chemoresistance linked to hSmo mutations. Guided by molecular docking and site-directed mutagenesis data, our work convincingly confirms that MRT-92 simultaneously recognized and occupied both sites 1 and 2. Our data demonstrate the existence of a third type of Smo antagonists, those entirely filling the Smo binding cavity from the upper extracellular part to the lower cytoplasmic-proximal subpocket. Our studies should help design novel potent Smo antagonists and more effective therapeutic strategies for treating Hh-linked cancers and associated chemoresistance.
Collapse
Affiliation(s)
- Lucile Hoch
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Helene Faure
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Hermine Roudaut
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Angele Schoenfelder
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Andre Mann
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Nicolas Girard
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Laure Bihannic
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Olivier Ayrault
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Elena Petricci
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Maurizio Taddei
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Didier Rognan
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Martial Ruat
- *Centre National de la Recherche Scientifique, Unité Mixte de Recherche-9197, Neuroscience Paris-Saclay Institute, Molecules Circuits Department, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-7200, Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Illkirch, France; Centre National de la Recherche Scientifique, Unité Mixte de Recherche-3306, Institut National de la Santé et de la Recherche Médicale U1005, Institut Curie, Centre Universitaire, Orsay, France; and Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| |
Collapse
|
17
|
Kwong L, Bijlsma MF, Roelink H. Shh-mediated degradation of Hhip allows cell autonomous and non-cell autonomous Shh signalling. Nat Commun 2014; 5:4849. [PMID: 25215859 PMCID: PMC4163939 DOI: 10.1038/ncomms5849] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022] Open
Abstract
The distribution of Sonic Hedgehog (Shh) is a highly regulated and critical process for development. Several negative feedback mechanisms are in place, including the Shh-induced upregulation of Hedgehog-interacting protein (Hhip). Hhip sequesters Shh, leading to a non-cell autonomous inhibition of the pathway. Hhip overexpression has a severe effect on neural tube development, raising the question why normal sites of Hhip expression have a seemingly unimpaired response to Shh. Here we show that although Hhip is able to leave its sites of synthesis to inhibit Shh non-cell autonomously, activation of Smoothened (Smo) drastically increases Hhip internalization and degradation cell autonomously. Although Hhip is unable to cell autonomously inhibit the consequences of Smo activation, it can inhibit the Shh response non-cell autonomously. Our data provide a mechanism by which the Shh ligand can activate the response and negate cell autonomous effects of Hhip, while Hhip can still induce non-cell autonomous inhibition.
Collapse
Affiliation(s)
- Lina Kwong
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, 3204, Berkeley, California 94720, USA
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Henk Roelink
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, 3204, Berkeley, California 94720, USA
| |
Collapse
|
18
|
El Waly B, Macchi M, Cayre M, Durbec P. Oligodendrogenesis in the normal and pathological central nervous system. Front Neurosci 2014; 8:145. [PMID: 24971048 PMCID: PMC4054666 DOI: 10.3389/fnins.2014.00145] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/23/2014] [Indexed: 12/26/2022] Open
Abstract
Oligodendrocytes (OLGs) are generated late in development and myelination is thus a tardive event in the brain developmental process. It is however maintained whole life long at lower rate, and myelin sheath is crucial for proper signal transmission and neuronal survival. Unfortunately, OLGs present a high susceptibility to oxidative stress, thus demyelination often takes place secondary to diverse brain lesions or pathologies. OLGs can also be the target of immune attacks, leading to primary demyelination lesions. Following oligodendrocytic death, spontaneous remyelination may occur to a certain extent. In this review, we will mainly focus on the adult brain and on the two main sources of progenitor cells that contribute to oligodendrogenesis: parenchymal oligodendrocyte precursor cells (OPCs) and subventricular zone (SVZ)-derived progenitors. We will shortly come back on the main steps of oligodendrogenesis in the postnatal and adult brain, and summarize the key factors involved in the determination of oligodendrocytic fate. We will then shed light on the main causes of demyelination in the adult brain and present the animal models that have been developed to get insight on the demyelination/remyelination process. Finally, we will synthetize the results of studies searching for factors able to modulate spontaneous myelin repair.
Collapse
Affiliation(s)
- Bilal El Waly
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Magali Macchi
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Myriam Cayre
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| | - Pascale Durbec
- CNRS, Institut de Biologie du Développement de Marseille UMR 7288, Aix Marseille Université Marseille, France
| |
Collapse
|
19
|
Yue S, Tang LY, Tang Y, Tang Y, Shen QH, Ding J, Chen Y, Zhang Z, Yu TT, Zhang YE, Cheng SY. Requirement of Smurf-mediated endocytosis of Patched1 in sonic hedgehog signal reception. eLife 2014; 3. [PMID: 24925320 PMCID: PMC4080449 DOI: 10.7554/elife.02555] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 06/10/2014] [Indexed: 01/17/2023] Open
Abstract
Cell surface reception of Sonic hedgehog (Shh) must ensure that the graded morphogenic signal is interpreted accordingly in neighboring cells to specify tissue patterns during development. Here, we report endocytic sorting signals for the receptor Patched1 (Ptch1), comprising two 'PPXY' motifs, that direct it to degradation in lysosomes. These signals are recognized by two HECT-domain ubiquitin E3 ligases, Smurf1 and Smurf2, which are induced by Shh and become enriched in Caveolin-1 lipid rafts in association with Ptch1. Smurf-mediated endocytic turnover of Ptch1 is essential for its clearance from the primary cilium and pathway activation. Removal of both Smurfs completely abolishes the ability of Shh to sustain the proliferation of postnatal granule cell precursors in the cerebellum. These findings reveal a novel step in the Shh pathway activation as part of the Ptch1 negative feedback loop that precisely controls the signaling output in response to Shh gradient signal.
Collapse
Affiliation(s)
- Shen Yue
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Liu-Ya Tang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, United States
| | - Ying Tang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Tang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, United States
| | - Qiu-Hong Shen
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jie Ding
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zengdi Zhang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Yu
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, United States
| | - Steven Y Cheng
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
20
|
Ruat M, Faure H, Daynac M. Smoothened, Stem Cell Maintenance and Brain Diseases. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_83] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
21
|
Abstract
The morphogen Sonic Hedgehog (Shh) controls the generation of oligodendrocyte (OLs) during embryonic development and regulates OL production in adulthood in the cortex and corpus callosum. The roles of Shh in CNS repair following lesions associated with demyelinating diseases are still unresolved. Here, we address this issue by using a model of focal demyelination induced by lysolecithin in the corpus callosum of adult mice. Shh transcripts and protein were not detected in control animals but were upregulated in a time-dependent manner in the oligodendroglial lineage within the lesion. We report an increased transcription of Shh target genes suggesting a broad reactivation of the Shh pathway. We show that the adenovirus-mediated transfer of Shh into the lesioned brain results in the attenuation of the lesion extent with the increase of OL progenitor cells (OPCs) and mature myelinating OL numbers due to survival, proliferation, and differentiation activities as well as the decrease of astrogliosis and macrophage infiltration. Furthermore, the blocking of Shh signaling during the lesion, using its physiological antagonist, Hedgehog interacting protein, results in a decrease of OPC proliferation and differentiation, preventing repair. Together, our findings identify Shh as a necessary factor playing a positive role during demyelination and indicate that its signaling activation stands as a potential therapeutic approach for myelin diseases.
Collapse
|
22
|
Angeloni N, Bond CW, Harrington D, Stupp S, Podlasek CA. Sonic hedgehog is neuroprotective in the cavernous nerve with crush injury. J Sex Med 2012; 10:1240-50. [PMID: 22994531 DOI: 10.1111/j.1743-6109.2012.02930.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION The cavernous nerve (CN) is commonly injured during prostatectomy, resulting in erectile dysfunction (ED). Although peripheral nerves have a limited ability to regenerate, a return of function typically does not occur due to irreversible down stream morphological changes in the penis that result from CN injury. We have shown in previous studies that sonic hedgehog (SHH) is critical for CN regeneration and improves erectile function after crush injury. AIMS Examine a new direction, to determine if SHH is neuroprotective to the pelvic ganglia (PG)/CN after crush injury. A secondary focus is to examine if SHH signaling decreases with age in the PG/CN. METHODS Sprague-Dawley rats underwent bilateral CN crush and SHH and glial fibrillary acidic protein were quantified by western analysis of the PG/CN (N = 6 rats at each time point) at 1, 2, 4, 7, and 14 days, and the apoptotic index was measured in the penis. SHH was quantified by western in the PG/CN with blockade of anterograde transport (N = 4 rats) in comparison to mouse IgG (N = 4 rats). If SHH is neuroprotective was examined at 4 (N = 14 rats) and 7 days (N = 16 rats) of treatment after CN crush. SHH protein was quantified in aging (P200-300, N = 5 rats) PG/CN in comparison to normal adult (P115-120, N = 3 rats) PG/CN. Main Outcome Measures. SHH pathway was examined in PG via immunohistochemistry, in situ, western, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). RESULTS SHH is neuroprotective in the PG/CN with injury. SHH localization in the PG/CN suggests SHH interaction in neuronal/glial signaling. SHH protein is significantly decreased in the PG/CN after crush injury and in the aged PG/CN. Signals from the PG are required to maintain SHH in the CN. CONCLUSIONS There is a window of opportunity immediately after nerve insult in which manipulation of SHH signaling in the nerve microenvironment can affect long-term regeneration outcome.
Collapse
Affiliation(s)
- Nicholas Angeloni
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
23
|
Estradiol triggers sonic-hedgehog-induced angiogenesis during peripheral nerve regeneration by downregulating hedgehog-interacting protein. J Transl Med 2012; 92:532-42. [PMID: 22330336 PMCID: PMC3319330 DOI: 10.1038/labinvest.2012.6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Both estradiol (E2) and Sonic Hedgehog (Shh) contribute to angiogenesis and nerve regeneration. Here, we investigated whether E2 improves the recovery of injured nerves by downregulating the Shh inhibitor hedgehog-interacting protein (HIP) and increasing Shh-induced angiogenesis. Mice were treated with local injections of E2 or placebo one week before nerve-crush injury; 28 days after injury, nerve conduction velocity, exercise duration, and vascularity were significantly greater in E2-treated mice than in placebo-treated mice. E2 treatment was also associated with higher mRNA levels of Shh, the Shh receptor Patched-1, and the Shh transcriptional target Gli1, but with lower levels of HIP. The E2-induced enhancement of nerve vascularity was abolished by the Shh inhibitor cyclopamine, and the effect of E2 treatment on Shh, Gli1, and HIP mRNA expression was abolished by the E2 inhibitor ICI. Gli-luciferase activity in human umbilical-vein endothelial cells (HUVECs) increased more after treatment with E2 and Shh than after treatment with E2 alone, and E2 treatment reduced HIP expression in HUVECs and Schwann cells without altering Shh expression. Collectively, these findings suggest that E2 improves nerve recovery, at least in part, by reducing HIP expression, which subsequently leads to an increase in Shh signaling and Shh-induced angiogenesis.
Collapse
|
24
|
Ruat M, Roudaut H, Ferent J, Traiffort E. Hedgehog trafficking, cilia and brain functions. Differentiation 2012; 83:S97-104. [DOI: 10.1016/j.diff.2011.11.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 11/21/2011] [Accepted: 11/22/2011] [Indexed: 10/14/2022]
|
25
|
Manetti F, Faure H, Roudaut H, Gorojankina T, Traiffort E, Schoenfelder A, Mann A, Solinas A, Taddei M, Ruat M. Virtual screening-based discovery and mechanistic characterization of the acylthiourea MRT-10 family as smoothened antagonists. Mol Pharmacol 2010; 78:658-65. [PMID: 20664000 DOI: 10.1124/mol.110.065102] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The seven-transmembrane receptor Smoothened (Smo) is the major component involved in signal transduction of the Hedgehog (Hh) morphogens. Smo inhibitors represent a promising alternative for the treatment of several types of cancers linked to abnormal Hh signaling. Here, on the basis of experimental data, we generated and validated a pharmacophoric model for Smo inhibitors constituted by three hydrogen bond acceptor groups and three hydrophobic regions. We used this model for the virtual screening of a library of commercially available compounds. Visual and structural criteria allowed the selection of 20 top scoring ligands, and an acylthiourea, N-(3-benzamidophenylcarbamothioyl)-3,4,5-trimethoxybenzamide (MRT-10), was identified and characterized as a Smo antagonist. The corresponding acylurea, N-(3-benzamidophenylcarbamoyl)-3,4,5-trimethoxybenzamide (MRT-14), was synthesized and shown to display, in various Hh assays, an inhibitory potency comparable to or greater than that of reference Smo antagonists cyclopamine and N-((3S,5S)-1-(benzo[d][1,3]dioxol-5-ylmethyl)-5-(piperazine-1-carbonyl)pyrrolidin-3-yl)-N-(3-methoxybenzyl)-3,3-dimethylbutanamide (Cur61414). Focused virtual screening of the same library further identified five additional related antagonists. MRT-10 and MRT-14 constitute the first members of novel families of Smo antagonists. The described virtual screening approach is aimed at identifying novel modulators of Smo and of other G-protein coupled receptors.
Collapse
Affiliation(s)
- Fabrizio Manetti
- Dipartimento Farmaco Chimico Tecnologico, Università degli Studi di Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Faure H, Gorojankina T, Rice N, Dauban P, Dodd RH, Bräuner-Osborne H, Rognan D, Ruat M. Molecular determinants of non-competitive antagonist binding to the mouse GPRC6A receptor. Cell Calcium 2010; 46:323-32. [PMID: 19836834 DOI: 10.1016/j.ceca.2009.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 09/16/2009] [Indexed: 10/20/2022]
Abstract
GPRC6A displays high sequence homology to the Ca2+-sensing receptor (CaSR). Here we report that the calcimimetic Calindol and the calcilytic NPS2143 antagonize increases in inositol phosphate elicited by L-ornithine-induced activation of mouse GPRC6A after transient coexpression with Galpha(qG66D) in HEK293 cells. The calcilytic Calhex 231 did not modulate this response. A three-dimensional model of the GPRC6A seven transmembrane domains (TMs) was constructed. It was used to identify seven residues strictly conserved within the CaSR and GPRC6A allosteric binding pockets, and previously demonstrated to interact with calcilytics or calcimimetics. The mutations F666A(3.32), F670A(3.36), W797A(6.48) caused a loss of L-ornithine ability to activate GPRC6A mutants. The F800A(6.51) mutant was not implicated in either Calindol or NPS 2143 recognition. The E816Q(7.39) mutation led to a loss of Calindol antagonist activity but was without effect on NPS2143 inhibitory response. In summary, these data suggest that Calindol is primarily anchored through an H-bond to E816(7.39) in TM7 and highlight important local differences at the level of the CaSR and GPRC6A allosteric binding pockets. We have identified the first antagonists of GPRC6A that could represent new tools to analyze GPRC6A functions and serve as chemical leads for the development of more specific modulators.
Collapse
Affiliation(s)
- Helene Faure
- CNRS, UPR9040, Institut de Neurobiologie Alfred Fessard-IFR 2118, Signal Transduction and Developmental Neuropharmacology Team, 1 Avenue de la Terrasse, F-91198 Gif-sur-Yvette, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The discovery of a Sonic Hedgehog (Shh) signaling pathway in the mature vertebrate CNS has paved the way to the characterization of the functional roles of Shh signals in normal and diseased brain. Shh is proposed to participate in the establishment and maintenance of adult neurogenic niches and to regulate the proliferation of neuronal or glial precursors in several brain areas. Consistent with its role during brain development, misregulation of Shh signaling is associated with tumorigenesis while its recruitement in damaged neural tissue might be part of the regenerating process. This review focuses on the most recent data of the Hedgehog pathway in the adult brain and its relevance as a novel therapeutic approach for brain diseases including brain tumors.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- CNRS, Alfred Fessard Institute of Neurobiology, Laboratory of Neurobiology and Development, UPR-3294, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France.
| | | | | |
Collapse
|
28
|
Sanek NA, Taylor AA, Nyholm MK, Grinblat Y. Zebrafish zic2a patterns the forebrain through modulation of Hedgehog-activated gene expression. Development 2009; 136:3791-800. [PMID: 19855021 PMCID: PMC2766342 DOI: 10.1242/dev.037820] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2009] [Indexed: 11/20/2022]
Abstract
Holoprosencephaly (HPE) is the most common congenital malformation of the forebrain in human. Several genes with essential roles during forebrain development have been identified because they cause HPE when mutated. Among these are genes that encode the secreted growth factor Sonic hedgehog (Shh) and the transcription factors Six3 and Zic2. In the mouse, Six3 and Shh activate each other's transcription, but a role for Zic2 in this interaction has not been tested. We demonstrate that in zebrafish, as in mouse, Hh signaling activates transcription of six3b in the developing forebrain. zic2a is also activated by Hh signaling, and represses six3b non-cell-autonomously, i.e. outside of its own expression domain, probably through limiting Hh signaling. Zic2a repression of six3b is essential for the correct formation of the prethalamus. The diencephalon-derived optic stalk (OS) and neural retina are also patterned in response to Hh signaling. We show that zebrafish Zic2a limits transcription of the Hh targets pax2a and fgf8a in the OS and retina. The effects of Zic2a depletion in the forebrain and in the OS and retina are rescued by blocking Hh signaling or by increasing levels of the Hh antagonist Hhip, suggesting that in both tissues Zic2a acts to attenuate the effects of Hh signaling. These data uncover a novel, essential role for Zic2a as a modulator of Hh-activated gene expression in the developing forebrain and advance our understanding of a key gene regulatory network that, when disrupted, causes HPE.
Collapse
Affiliation(s)
- Nicholas A Sanek
- Department of Zoology and Anatomy, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
29
|
Angeloni NL, Bond CW, Monsivais D, Tang Y, Podlasek CA. The role of hedgehog-interacting protein in maintaining cavernous nerve integrity and adult penile morphology. J Sex Med 2009; 6:2480-93. [PMID: 19515211 PMCID: PMC2814768 DOI: 10.1111/j.1743-6109.2009.01349.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Sonic hedgehog (SHH) is an essential regulator of smooth muscle apoptosis in the penis that has significant clinical potential as a therapy to suppress post-prostatectomy apoptosis, an underlying cause of erectile dysfunction (ED). Thus an understanding of how SHH signaling is regulated in the adult penis is essential to move the field of ED research forward and to develop new treatment strategies. We propose that hedgehog-interacting protein (HIP), which has been shown to bind SHH protein and to play a role in SHH regulation during embryogenesis of other organs, is a critical regulator of SHH signaling, penile morphology, and apoptosis induction. AIMS We have examined HIP signaling in the penis and cavernous nerve (CN) during postnatal differentiation of the penis, in CN-injured, and a diabetic model of ED. METHODS HIP localization/abundance and RNA abundance were examined by immunohistochemical (IHC) analysis and real-time reverse transcriptase-polymerase chain reaction (RT-PCR) in Sprague-Dawley rats between the ages of 7 and 92 days old, in CN-injured Sprague-Dawley rats and in BioBreeding/Worcester diabetic rats. HIP signaling was perturbed in the pelvic ganglia and in the penis and TUNEL assay was performed in the penis. CN tie, lidocaine, and anti-kinesin experiments were performed to examine HIP signaling in the CN and penis. RESULTS In this study we are the first to demonstrate that HIP undergoes anterograde transport to the penis via the CN, that HIP perturbation in the pelvic ganglia or the penis induces apoptosis, and that HIP plays a role in maintaining CN integrity, penile morphology, and SHH abundance. CONCLUSIONS These studies are significant because they show HIP involvement in cross-talk (signaling) between the pelvic ganglia and penis, which is integral for maintenance of penile morphology and they suggest a mechanism of how nerves may regulate target organ morphology and function.
Collapse
Affiliation(s)
- Nicholas L Angeloni
- Department of Urology, Northwestern University Medical School, Chicago, IL 60611, USA
| | | | | | | | | |
Collapse
|
30
|
Ribes V, Briscoe J. Establishing and interpreting graded Sonic Hedgehog signaling during vertebrate neural tube patterning: the role of negative feedback. Cold Spring Harb Perspect Biol 2009; 1:a002014. [PMID: 20066087 PMCID: PMC2742090 DOI: 10.1101/cshperspect.a002014] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The secreted protein Sonic Hedgehog (SHH) acts in graded fashion to pattern the dorsal-ventral axis of the vertebrate neural tube. This is a dynamic process in which increasing concentrations and durations of exposure to SHH generate neurons with successively more ventral identities. Interactions between the receiving cells and the graded signal underpin the mechanism of SHH action. In particular, negative feedback, involving proteins transcriptionally induced or repressed by SHH signaling, plays an essential role in shaping the graded readout. On one hand, negative feedback controls, in a noncell-autonomous manner, the distribution of SHH across the field of receiving cells. On the other, it acts cell-autonomously to convert different concentrations of SHH into distinct durations of intracellular signal transduction. Together, these mechanisms exemplify a strategy for morphogen interpretation, which we have termed temporal adaptation that relies on the continuous processing and refinement of the cellular response to the graded signal.
Collapse
Affiliation(s)
| | - James Briscoe
- Developmental Neurobiology, National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom, NW7 1AA
| |
Collapse
|
31
|
Structural insights into hedgehog ligand sequestration by the human hedgehog-interacting protein HHIP. Nat Struct Mol Biol 2009; 16:698-703. [PMID: 19561611 PMCID: PMC2709225 DOI: 10.1038/nsmb.1607] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 04/17/2009] [Indexed: 12/21/2022]
Abstract
Hedgehog (Hh) morphogens have fundamental roles in development, whereas dysregulation of Hh signaling leads to disease. Multiple cell-surface receptors are responsible for transducing and/or regulating Hh signals. Among these, the Hedgehog-interacting protein (Hhip) is a highly conserved, vertebrate-specific inhibitor of Hh signaling. We have solved a series of crystal structures for the human HHIP ectodomain and Desert hedgehog (DHH) in isolation, as well as HHIP in complex with DHH (HHIP-DHH) and Sonic hedgehog (Shh) (HHIP-Shh), with and without Ca2+. The interaction determinants, confirmed by biophysical studies and mutagenesis, reveal previously uncharacterized and distinct functions for the Hh Zn2+ and Ca2+ binding sites--functions that may be common to all vertebrate Hh proteins. Zn2+ makes a key contribution to the Hh-HHIP interface, whereas Ca2+ is likely to prevent electrostatic repulsion between the two proteins, suggesting an important modulatory role. This interplay of several metal binding sites suggests a tuneable mechanism for regulation of Hh signaling.
Collapse
|
32
|
Hasebe T, Kajita M, Shi YB, Ishizuya-Oka A. Thyroid hormone-up-regulated hedgehog interacting protein is involved in larval-to-adult intestinal remodeling by regulating sonic hedgehog signaling pathway in Xenopus laevis. Dev Dyn 2008; 237:3006-15. [PMID: 18816855 DOI: 10.1002/dvdy.21698] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Sonic hedgehog (Shh) was previously shown to be involved in the larval-to-adult remodeling of the Xenopus laevis intestine. While Shh is transcriptionally regulated by thyroid hormone (TH), the posttranscriptional regulation of Shh signaling during intestinal remodeling is largely unknown. In the present study, we focused on a role of the pan-hedgehog inhibitor, hedgehog interacting protein (Hip), in the spatiotemporal regulation of Shh signaling. Using real-time reverse transcriptase-polymerase chain reaction and in situ hybridization, we show that Hip expression is transiently up-regulated during both natural and TH-induced metamorphosis and that Hip mRNA is localized in the connective tissue adjacent to the adult epithelial primordia expressing Shh. Interestingly, the expression of bone morphogenetic protein-4, a Shh target gene, is hardly detectable where Hip is strongly expressed. Finally, we demonstrate that Hip binds to the N-terminal fragment of processed Shh in vivo, suggesting that Hip suppresses Shh signaling through sequestering Shh.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, Nakahara-ku, Kawasaki, Kanagawa, Japan.
| | | | | | | |
Collapse
|
33
|
Chemoattractive Activity of Sonic Hedgehog in the Adult Subventricular Zone Modulates the Number of Neural Precursors Reaching the Olfactory Bulb. Stem Cells 2008; 26:2311-20. [DOI: 10.1634/stemcells.2008-0297] [Citation(s) in RCA: 340] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
34
|
Abstract
Most of the previous work on the sphingolipid ceramide has been devoted to its function as an apoptosis inducer. Recent studies, however, have shown that in stem cells, ceramide has additional nonapoptotic functions. In this article, ceramide signaling will be reviewed in light of 'systems interface biology': as an interconnection of sphingolipid metabolism, membrane biophysics and cell signaling. The focus will be on the metabolic interconversion of ceramide and sphingomyelin or sphingosine-1-phosphate. Lipid rafts and sphingolipid-induced protein scaffolds will be discussed as a membrane interface for lipid-controlled cell signaling. Ceramide/sphingomyelin and ceramide/sphingosine-1-phosphate-interdependent cell-signaling pathways are significant for the regulation of cell polarity, apoptosis and/or proliferation, and as novel pharmacologic targets in cancer and stem cells.
Collapse
Affiliation(s)
- Erhard Bieberich
- Institute of Molecular Medicine & Genetics, School of Medicine, Medical College of Georgia, 1120 15th Street, Room CB-2803, Augusta, GA 30912, USA
| |
Collapse
|
35
|
Jozwiak J, Grajkowska W, Wlodarski P. Pathogenesis of medulloblastoma and current treatment outlook. Med Res Rev 2008; 27:869-90. [PMID: 17089411 DOI: 10.1002/med.20088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Medulloblastoma is the most common malignant tumor of the cerebellum in children, with a tendency to metastasize via CSF pathway. Survival rate varies depending on several factors, but is rather favorable, with radiotherapy as the treatment of choice. Irradiation of the craniospinal axis results, however, in severe neuropsychological and psychosocial impairments pertaining to memory, attention, motor functioning, language, and visuospatial abilities. Precise mechanisms underlying the formation of medulloblastoma are still unclear, but implication of at least three signaling molecules is postulated: insulin-like growth factor-I, WNT, and Sonic hedgehog. Thanks to increasing knowledge on the cellular mechanisms contributing to tumor formation, it is possible to propose new therapies that could replace radiotherapy or allow decreasing irradiation doses. The current review presents recent developments in medulloblastoma pathophysiology research and proposed inhibitors that could constitute good candidates for further pharmacological research.
Collapse
Affiliation(s)
- Jaroslaw Jozwiak
- Department of Histology and Embryology, Center for Biostructure Research, Medical University of Warsaw, Warsaw, Poland.
| | | | | |
Collapse
|
36
|
Recent progress in the study of Hedgehog signaling. J Genet Genomics 2008; 35:129-37. [DOI: 10.1016/s1673-8527(08)60019-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Revised: 11/20/2007] [Accepted: 11/21/2007] [Indexed: 12/30/2022]
|
37
|
Masdeu C, Bernard V, Faure H, Traiffort E, Ruat M. Distribution of Smoothened at hippocampal mossy fiber synapses. Neuroreport 2007; 18:395-9. [PMID: 17435610 DOI: 10.1097/wnr.0b013e32801421ce] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The seven-transmembrane receptor Smoothened is essential for hedgehog signal transduction. In adulthood, the highest density of Smoothened mRNA is found in the granule cell layer of the dentate gyrus. There, Smoothened expression is regulated by the synaptic activity involving the glutamatergic transmission. The precise localization of Smoothened proteins, however, has not yet been reported. Here, we describe Smoothened protein distribution in the hippocampal mossy fibers using specific Smoothened antibodies. We provide evidences for their presynaptic localization, and using electron microscopy, show that Smoothened is located in close association with synaptic vesicles and rarely with the plasma membrane. These findings demonstrate that Smoothened is localized presynaptically and suggest that Smoothened signal transduction may be implicated in the complex aspects of mossy fiber function.
Collapse
Affiliation(s)
- Christelle Masdeu
- Laboratory of Cellular and Molecular Neurobiology, Neurobiology Institute Alfred Fessard-IFR 2118, Signal Transduction and Developmental Neuropharmacology Team, Gif-sur-Yvette, France
| | | | | | | | | |
Collapse
|
38
|
Abstract
Locomotion mediated by skeletal muscle provides a basis for the behavioral repertoire of most animals. Embryological and genetic studies of mouse, bird, fish and frog embryos are providing insights into the functions of the myogenic regulatory factors (MRFs) and the signaling molecules that regulate activity of MRFs. Nevertheless, our understanding of muscle development remains somewhat limited. Fundamental goals are to elucidate how mesodermal cells are induced during gastrulation to form muscle precursor cells and how muscle precursor cells acquire specific cell fates, such as slow and fast muscle cells. In this review, we focus on studies of zebrafish muscle development that have advanced our understanding of the molecular genetics of muscle cell induction and specification.
Collapse
Affiliation(s)
- Haruki Ochi
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403-1254, USA
| | | |
Collapse
|
39
|
Abstract
Hedgehog signaling coordinates a variety of patterning processes during early embryonic development. Drosophila hedgehog and its vertebrate orthologs, Sonic hedgehog, Indian hedgehog, and Desert hedgehog, share a generally conserved signal transduction cascade. However, the particular mechanisms by which the lipid-modified molecules specify embryonic tissues differ substantially. Vertebrate skeletal patterning is one of the most intensively studied biological processes. During skeletogenesis, Sonic and Indian hedgehog provide positional information and initiate or maintain cellular differentiation programs regulating the formation of cartilage and bone. They either signal directly to adjacent cells or form tightly regulated gradients that act over long distances to pattern the axial and appendicular skeleton and regulate crucial steps during endochondral ossification. As a consequence, malfunction of the hedgehog signaling network can cause severe skeletal disorders and tumors.
Collapse
Affiliation(s)
- Harald W A Ehlen
- University of Duisburg-Essen, Center for Medical Biotechnology, Essen, Germany
| | | | | |
Collapse
|
40
|
Masdeu C, Faure H, Coulombe J, Schoenfelder A, Mann A, Brabet I, Pin JP, Traiffort E, Ruat M. Identification and characterization of Hedgehog modulator properties after functional coupling of Smoothened to G15. Biochem Biophys Res Commun 2006; 349:471-9. [PMID: 16945339 DOI: 10.1016/j.bbrc.2006.07.216] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 07/28/2006] [Indexed: 11/17/2022]
Abstract
The seven-transmembrane receptor Smoothened (Smo) transduces the signal initiated by Hedgehog (Hh) morphogen binding to the receptor Patched (Ptc). We have reinvestigated the pharmacological properties of reference molecules acting on the Hh pathway using various Hh responses and a novel functional assay based on the coexpression of Smo with the alpha subunit of the G15 protein in HEK293 cells. The measurement of inositol phosphate (IP) accumulation shows that Smo has constitutive activity, a response blocked by Ptc which indicates a functional Hh receptor complex. Interestingly, the antagonists cyclopamine, Cur61414, and SANT-1 display inverse agonist properties and the agonist SAG has no effect at the Smo-induced IP response, but converts Ptc-mediated inactive forms of Smo into active ones. An oncogenic Smo mutant does not mediate an increase in IP response, presumably reflecting its inability to reach the cell membrane. These studies identify novel properties of molecules displaying potential interest in the treatment of various cancers and brain diseases, and demonstrate that Smo is capable of signaling through G15.
Collapse
Affiliation(s)
- Christelle Masdeu
- CNRS, Institut de Neurobiologie Alfred Fessard-IFR 2118, UPR9040, Laboratoire de Neurobiologie Cellulaire et Moléculaire, Signal Transduction and Developmental Neuropharmacology team, 1 avenue de la Terrasse, Gif-sur-Yvette F-91198, France
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The hedgehog pathway is a major regulator of embryonic development, and mutations that decrease its activity are known to be associated with severe defects in nervous system development. Recent evidence suggests hedgehog continues to function in adult tissue, normal as well as diseased, by regulating both cell proliferation and the production of growth and angiogenic factors. In the adult nervous system, this dual ability is especially important in regulating the behavior of neural stem and progenitor cells. This review summarizes information connecting hedgehog signaling and neural diseases, including neurodegenerative disorders and brain tumors, particularly medulloblastoma. We also describe the discovery and utility of small molecule agonists and antagonists of this pathway and their potential as novel types of therapeutics.
Collapse
|
42
|
Loulier K, Ruat M, Traiffort E. Increase of proliferating oligodendroglial progenitors in the adult mouse brain upon Sonic hedgehog delivery in the lateral ventricle. J Neurochem 2006; 98:530-42. [PMID: 16805844 DOI: 10.1111/j.1471-4159.2006.03896.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Sonic hedgehog signaling is required for the maintenance of stem cell niches in the postnatal subventricular zone and the proliferation of neural progenitors in the mature hippocampus. We show here that delivery of Sonic hedgehog protein into the lateral ventricle of adult mice increases cell proliferation in the corpus callosum and cerebral cortex. In this latter area, the number of neural progenitors expressing the proteoglycan NG2 is enhanced 2 days after the injection. In both areas, mRNA up-regulation of the transcriptional target gene Patched was observed in cells expressing the oligodendroglial transcription factor Olig1. Twenty-six days following the adenovirus-mediated delivery of Sonic hedgehog into the lateral ventricle, newly generated cells in the cerebral cortex and in the corpus callosum are influenced towards the initial steps of oligodendrogenesis, as indicated by a 50% increase in the number of cells expressing the oligodendroglial marker DM20. Our experiments demonstrate that the number of oligodendrocyte precursor cells in the cerebral cortex and corpus callosum can be increased upon delivery of Sonic hedgehog proteins and highlight the potential capacity of the adult brain to mobilize a pool of premyelinating cells.
Collapse
Affiliation(s)
- Karine Loulier
- CNRS, Signal Transduction and Developmental Neuropharmacology, UPR9040 Laboratoire de Neurobiologie Cellulaire et Moléculaire, Institut de Neurobiologie Alfred Fessard, IFR 2118, Gif sur Yvette, France
| | | | | |
Collapse
|
43
|
Ochi H, Pearson BJ, Chuang PT, Hammerschmidt M, Westerfield M. Hhip regulates zebrafish muscle development by both sequestering Hedgehog and modulating localization of Smoothened. Dev Biol 2006; 297:127-40. [PMID: 16765934 DOI: 10.1016/j.ydbio.2006.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 04/22/2006] [Accepted: 05/03/2006] [Indexed: 11/29/2022]
Abstract
Sharp borders between cells with different developmental fates are important for patterning of invertebrates, but are not well understood in vertebrates. Zebrafish slow muscle cells develop from adaxial cells, a one-cell-diameter-thick pseudo-epithelium immediately adjacent to the notochord. Hedgehog (Hh) signals from notochord specify adaxial cells to form slow muscle cells. Cells next to adaxial cells form fast muscle. This suggests that Hh signaling is locally regulated to produce a sharp border that separates slow and fast muscle precursors. To understand how Hh activity is locally regulated, we characterized the dynamic roles of Hhip, a protein that binds Hedgehog at the cell surface. Hhip is strongly expressed by adaxial cells and, together with Patched, the Hedgehog receptor, limits transduction of the Hedgehog signaling by Smoothened to adaxial cells. Hhip protein lacking its membrane associated domain still suppresses Hh activity but no longer acts synergistically with Patched. Hhip and Smoothened colocalize at the cell surface and, in response to Hedgehog, internalize together. Knockdown of Hhip blocks Smoothened internalization while increasing Hedgehog signaling and slow muscle formation. These data support a model in which Hhip regulates muscle development both by sequestering Hedgehog and by modulating localization of Smoothened.
Collapse
Affiliation(s)
- Haruki Ochi
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403-1254, USA
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Abstract
During development, secreted signaling factors, called morphogens, instruct cells to adopt specific mature phenotypes. However, the mechanisms that morphogen systems employ to establish a precise concentration gradient for patterning tissue architecture are highly complex and are typically analyzed only at long times after secretion (i.e. steady state). We have developed a theoretical model that analyzes dynamically how the intricate transport and signal transduction mechanisms of a model morphogen, Sonic hedgehog (Shh),cooperate in modular fashion to regulate tissue patterning in the neural tube. Consistent with numerous recent studies, the model elucidates how the dynamics of gradient formation can be a key determinant of cell response. In addition,this work yields several novel insights into how different transport mechanisms or `modules' control pattern formation. The model predicts that slowing the transport of a morphogen, such as by lipid modification of the ligand Shh, by ligand binding to proteoglycans, or by the moderate upregulation of dedicated transport molecules like Dispatched, can actually increase the signaling range of the morphogen by concentrating it near the secretion source. Furthermore, several transcriptional targets of Shh, such as Patched and Hedgehog-interacting protein, significantly limit its signaling range by slowing transport and promoting ligand degradation. This modeling approach elucidates how individual modular elements that operate dynamically at various times during patterning can shape a tissue pattern.
Collapse
Affiliation(s)
- Krishanu Saha
- Department of Chemical Engineering and the Helen Wills Neuroscience Institute, University of California, Berkeley, 94720-1462, USA
| | | |
Collapse
|
46
|
Loulier K, Ruat M, Traiffort E. Analysis of hedgehog interacting protein in the brain and its expression in nitric oxide synthase-positive cells. Neuroreport 2005; 16:1959-62. [PMID: 16272887 DOI: 10.1097/01.wnr.0000187632.91375.81] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hedgehog interacting protein (Hip) and Patched 1 (Ptc1) regulate the cell responses to the morphogen Sonic Hedgehog (Shh). Here, we compare the relative expression patterns of Shh, Hip and Ptc1 transcripts in the E13.5 mouse brain embryo. We observe that the expression of Hip and Ptc1 often overlaps and is found close to Shh-expressing cells, suggesting that both proteins are required for controlling Shh signals. In the adult striatum in which Ptc1 is not detected, we show that a majority of Hip-expressing cells correspond to neurons expressing the neuronal form of nitric oxide synthase. These data raise the hypothesis for a functional link between nitric oxide and Shh signaling and for a nonredundant role of Hip and Ptc1 in the adult brain.
Collapse
Affiliation(s)
- Karine Loulier
- CNRS, Signal Transduction and Developmental Neuropharmacology, Laboratory of Cellular and Molecular Neurobiology, Alfred Fessard Institute of Neurobiology, Gif sur Yvette, France
| | | | | |
Collapse
|
47
|
Torroja C, Gorfinkiel N, Guerrero I. Mechanisms of Hedgehog gradient formation and interpretation. ACTA ACUST UNITED AC 2005; 64:334-56. [PMID: 16041759 DOI: 10.1002/neu.20168] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Morphogens are molecules that spread from localized sites of production, specifying distinct cell outcomes at different concentrations. Members of the Hedgehog (Hh) family of signaling molecules act as morphogens in different developmental systems. If we are to understand how Hh elicits multiple responses in a temporally and spatially specific manner, the molecular mechanism of Hh gradient formation needs to be established. Moreover, understanding the mechanisms of Hh signaling is a central issue in biology, not only because of the role of Hh in morphogenesis, but also because of its involvement in a wide range of human diseases. Here, we review the mechanisms affecting the dynamics of Hh gradient formation, mostly in the context of Drosophila wing development, although parallel findings in vertebrate systems are also discussed.
Collapse
Affiliation(s)
- Carlos Torroja
- Centro de Biología Molecular Severo Ochoa, CSIC, Universidad Autónoma de Madrid, Cantoblanco, E-28049 Madrid, Spain
| | | | | |
Collapse
|
48
|
Pascual O, Traiffort E, Baker DP, Galdes A, Ruat M, Champagnat J. Sonic hedgehog signalling in neurons of adult ventrolateral nucleus tractus solitarius. Eur J Neurosci 2005; 22:389-96. [PMID: 16045492 DOI: 10.1111/j.1460-9568.2005.04223.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The transmembrane receptor Patched (Ptc) mediates the action of the diffusing factor Sonic hedgehog (Shh), which is implicated in establishing morphogenetic gradients during embryonic development. Whereas alteration of Ptc function is associated with developmental abnormalities and brain tumors, its functional activity and roles in the adult brain have yet to be elucidated. Here we describe the complementary pattern of Shh and Ptc expression in the rat dorsal vagal motor nucleus and the ventrolateral nucleus tractus solitarius (vNTS), respectively. Those two interconnected structures regulate the cardiorespiratory function during hypoxia. Bath application of a subnanomolar concentration of aminoterminal Shh protein (ShhN) to a slice preparation of the vNTS induces a rapid decrease in neuronal firing followed by a bursting activity that propagates in the neuronal network. Intracellular current injections show that bursts result from an action on the neuronal membrane electro-responsiveness. Both inhibiting and bursting effects are blocked by the monoclonal Shh antibody 5E1 and may require the Ptc binding site of ShhN. Thus, ShhN acting on specific neuronal sites controls electrophysiological properties of differentiated neurons of the vNTS. We speculate on a retrocontrol of cardiorespiratory signals in the vNTS, by Shh generated in dorsal vagal motoneurons.
Collapse
Affiliation(s)
- Olivier Pascual
- Neurobiologie Génétique et Intégrative, UPR 2216-CNRS, Institute de Neurobiologie Alfred Fessard, IFR 2118-CNRS, 1, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The Hedgehog (Hh)-signaling pathway is essential for numerous developmental processes in Drosophila and vertebrate embryos. Hh signal transduction encompasses a complex series of regulatory events, including the generation of the mature Hh ligand, propagation of the ligand from source of production as well as the reception and interpretation of the signal in Hh-receiving cells. Many congenital malformations in humans are known to involve mutations in various components of the Hh-signaling pathway. This mini review summarizes some recent findings about the regulation of Hh signal transduction and describes the spectrum of human congenital malformations that are associated with aberrant Hh signaling. Based on a comparison of mouse-mutant phenotypes and human syndromes, we discuss how Hh-dependent Gli activator and repressor functions contribute to some of the congenital malformations.
Collapse
Affiliation(s)
- E Nieuwenhuis
- Program in Developmental Biology, The Hospital for Sick Children, Department of Molecular and Medical Genetics, University of Toronto, Toronto, Ontario MG5 1X8, Canada
| | | |
Collapse
|
50
|
Traiffort E, Dubourg C, Faure H, Rognan D, Odent S, Durou MR, David V, Ruat M. Functional characterization of sonic hedgehog mutations associated with holoprosencephaly. J Biol Chem 2004; 279:42889-97. [PMID: 15292211 DOI: 10.1074/jbc.m405161200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations of the developmental gene Sonic hedgehog (SHH) and alterations of SHH signaling have been associated with holoprosencephaly (HPE), a rare disorder characterized by a large spectrum of brain and craniofacial anomalies. Based on the crystal structure of mouse N-terminal and Drosophila C-terminal hedgehog proteins, we have developed three-dimensional models of the corresponding human proteins (SHH-N, SHH-C) that have allowed us to identify within these two domains crucial regions associated with HPE missense mutations. We have further characterized the functional consequences linked to 11 of these mutations. In transfected HEK293 cells, the production of the active SHH-N fragment was dramatically impaired for eight mutants (W117R, W117G, H140P, T150R, C183F, L271P, I354T, A383T). The supernatants from these cell cultures showed no significant SHH-signaling activity in a reporter cell-based assay. Two mutants (G31R, D222N) were associated with a lower production of SHH-N and signaling activity. Finally, one mutant harboring the A226T mutation displays an activity comparable with the wild-type protein. This work demonstrates that most of the HPE-associated SHH mutations analyzed have a deleterious effect on the availability of SHH-N and its biological activity. However, because of the lack of correlation between genotype and phenotype for SHH-associated mutations, our study suggests that other factors intervene in the development of the spectrum of HPE anomalies.
Collapse
Affiliation(s)
- Elisabeth Traiffort
- Institut de Neurobiologie Alfred Fessard, IFR 2118 CNRS, Laboratoire de Neurobiologie Cellulaire et Moléculaire, UPR 9040 CNRS, Bātiment 33, 1 avenue de la terrasse 91198 Gif sur Yvette, France.
| | | | | | | | | | | | | | | |
Collapse
|