1
|
Lue JC, Radisky DC. From Embryogenesis to Senescence: The Role of Mammary Gland Physiology in Breast Cancer Risk. Cancers (Basel) 2025; 17:787. [PMID: 40075637 PMCID: PMC11898936 DOI: 10.3390/cancers17050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The mammary gland undergoes significant changes throughout a woman's life; from embryonic development to transformations after breastfeeding and during aging. These processes, while essential for normal breast physiology, can increase breast cancer risk when disrupted. This review explores three critical stages: embryonic development; postlactational involution; and age-related lobular involution (ARLI). We highlight key signaling pathways-Wnt, FGF, SHH, Notch, EGFR, and BMP-that guide embryonic development and discuss how their dysregulation can contribute to abnormal growth. For postlactational involution, we examine the two-phase process of cell death and tissue remodeling, showing how disruptions during this period, particularly postpartum, may foster a tumor-promoting environment. We also delve into ARLI and the role of cellular senescence in the aging mammary gland, focusing on the senescence-associated secretory phenotype (SASP) and its impact on inflammation and tissue remodeling. Understanding these processes provides new opportunities for breast cancer prevention and treatment strategies.
Collapse
Affiliation(s)
- Jaida C. Lue
- Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Derek C. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
2
|
Hu Y, Peng L, Zhuo X, Yang C, Zhang Y. Hedgehog Signaling Pathway in Fibrosis and Targeted Therapies. Biomolecules 2024; 14:1485. [PMID: 39766192 PMCID: PMC11727624 DOI: 10.3390/biom14121485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 01/12/2025] Open
Abstract
Hedgehog (Hh) signaling is a well-established developmental pathway; it is crucial for early embryogenesis, cell differentiation, and damage-driven regeneration. It is being increasingly recognized that dysregulated Hh signaling is also involved in fibrotic diseases, which are characterized by excessive extracellular matrix deposition that compromises tissue architecture and function. As in-depth insights into the mechanisms of Hh signaling are obtained, its complex involvement in fibrosis is gradually being illuminated. Notably, some Hh-targeted inhibitors are currently under exploration in preclinical and clinical trials as a means to prevent fibrosis progression. In this review, we provide a concise overview of the biological mechanisms involved in Hh signaling. We summarize the latest advances in our understanding of the roles of Hh signaling in fibrogenesis across the liver, kidneys, airways, and lungs, as well as other tissues and organs, with an emphasis on both the shared features and, more critically, the distinct functional variations observed across these tissues and organs. We thus highlight the context dependence of Hh signaling, as well as discuss the current status and the challenges of Hh-targeted therapies for fibrosis.
Collapse
Affiliation(s)
- Yuchen Hu
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linrui Peng
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhuo
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China;
| | - Yuwei Zhang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Wang T, Wang S, Wang T, Jia L, Nan G, Wang L. Cdc14B/Cyclin B1 signaling modulates the pathogenesis of sonic hedgehog subtype medulloblastoma. Am J Cancer Res 2024; 14:2868-2880. [PMID: 39005661 PMCID: PMC11236779 DOI: 10.62347/cvay8707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 05/13/2024] [Indexed: 07/16/2024] Open
Abstract
Medulloblastoma (MB) is a severe malignancy of the central nervous system that predominantly occurs in the cerebellum of children. Overactivation of the sonic hedgehog (Shh) signaling pathway is the primary cause of the development and progression of Shh subtype MB, although the detailed mechanisms underlying this process remain largely elusive. In this study, we discovered that Shh can promote proliferation in MB cells through non-canonical Hedgehog signaling. This involves Shh binding to Patched 1, disrupting its interaction with Cyclin B1, allowing for nuclear translocation of Cyclin B1, and inducing the activation of genes involved in cell division. Furthermore, we observed that deregulation of Cdc14B leads to the stabilization of the Cyclin B1/CDK1 complex in MB cells through activating Cdc25C, a phosphatase known to help maintain Cyclin B1 stability. Our findings highlight the role of Cdc14B/Cdc25C/CDK1/Cyclin B1 in mediating Hedgehog signaling-driven pathogenesis in MB and have implications for identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Shan Wang
- Institute of Basic Translational Medicine, Xi’an Medical UniversityXi’an 710021, Shaanxi, China
| | - Tao Wang
- The No. 2 Department of Neurology, Shaanxi Province People’s HospitalXi’an 710068, Shaanxi, China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Gang Nan
- Department of Cell Biology, School of Basic Medical Sciences & National Translational Science Center for Molecular Medicine, Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical UniversityXi’an 710038, Shaanxi, China
| |
Collapse
|
4
|
Jeng KS, Chang CF, Tsang YM, Sheen IS, Jeng CJ. Reappraisal of the Roles of the Sonic Hedgehog Signaling Pathway in Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:1739. [PMID: 38730691 PMCID: PMC11083695 DOI: 10.3390/cancers16091739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
HCC remains one of the leading causes of cancer-related death globally. The main challenges in treatments of hepatocellular carcinoma (HCC) primarily arise from high rates of postoperative recurrence and the limited efficacy in treating advanced-stage patients. Various signaling pathways involved in HCC have been reported. Among them, the Sonic hedgehog (SHH) signaling pathway is crucial. The presence of SHH ligands is identified in approximately 60% of HCC tumor tissues, including tumor nests. PTCH-1 and GLI-1 are detected in more than half of HCC tissues, while GLI-2 is found in over 84% of HCC tissues. The SHH signaling pathway (including canonical and non-canonical) is involved in different aspects of HCC, including hepatocarcinogenesis, tumor growth, tumor invasiveness, progression, and migration. The SHH signaling pathway also contributes to recurrence, metastasis, modulation of the cancer microenvironment, and sustaining cancer stem cells. It also affects the resistance of HCC cells to chemotherapy, target therapy, and radiotherapy. Reappraisal of the roles of the SHH signaling pathway in HCC may trigger some novel therapies for HCC.
Collapse
Affiliation(s)
- Kuo-Shyang Jeng
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Chiung-Fang Chang
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - Yuk-Ming Tsang
- Department of Imaging Medicine, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - I-Shyan Sheen
- Department of Gastroenterology & Hepatology, Linkou Chang Memorial Hospital, Chang Gung Medical Foundation, Taoyuan City 333, Taiwan;
| | - Chi-Juei Jeng
- Graduate Institude of Clinical Medicine, National Taiwan University, College of Medicine, Taipei City 10617, Taiwan;
| |
Collapse
|
5
|
Singh R, Ray A. Therapeutic potential of hedgehog signaling in advanced cancer types. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:49-80. [PMID: 38782501 DOI: 10.1016/bs.ircmb.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
In this chapter, we have made an attempt to elucidate the relevance of hedgehog signaling pathway in tumorigenesis. Here, we have described different types of hedgehog signaling (canonical and non-canonical) with emphasis on the different mechanisms (mutation-driven, autocrine, paracrine and reverse paracrine) it adopts during tumorigenesis. We have discussed the role of hedgehog signaling in regulating cell proliferation, invasion and epithelial-to-mesenchymal transition in both local and advanced cancer types, as reported in different studies based on preclinical and clinical models. We have specifically addressed the role of hedgehog signaling in aggressive neuroendocrine tumors as well. We have also elaborated on the studies showing therapeutic relevance of the inhibitors of hedgehog signaling in cancer. Evidence of the crosstalk of hedgehog signaling components with other signaling pathways and treatment resistance due to tumor heterogeneity have also been briefly discussed. Together, we have tried to put forward a compilation of the studies on therapeutic potential of hedgehog signaling in various cancers, specifically aggressive tumor types with a perspective into what is lacking and demands further investigation.
Collapse
Affiliation(s)
- Richa Singh
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States.
| | - Anindita Ray
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| |
Collapse
|
6
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
7
|
Xiao Y, Chen X, Hu W, Ma W, Di Q, Tang H, Zhao X, Huang G, Chen W. USP39-mediated deubiquitination of Cyclin B1 promotes tumor cell proliferation and glioma progression. Transl Oncol 2023; 34:101713. [PMID: 37302347 DOI: 10.1016/j.tranon.2023.101713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND The elevated Cyclin B1 expression contributes to various tumorigenesis and poor prognosis. Cyclin B1 expression could be regulated by ubiquitination and deubiquitination. However, the mechanism of how Cyclin B1 is deubiquitinated and its roles in human glioma remain unclear. METHODS Co-immunoprecipitation and other assays were performed to detect the interacting of Cyclin B1 and USP39. A series of in vitro and in vivo experiments were performed to investigate the effect of USP39 on the tumorigenicity of tumor cells. RESULTS USP39 interacts with Cyclin B1 and stabilizes its expression by deubiquitinating Cyclin B1. Notably, USP39 cleaves the K29-linked polyubiquitin chain on Cyclin B1 at Lys242. Additionally, overexpression of Cyclin B1 rescues the arrested cell cycle at G2/M transition and the suppressed proliferation of glioma cells caused by USP39 knockdown in vitro. Furthermore, USP39 promotes the growth of glioma xenograft in subcutaneous and in situ of nude mice. Finally, in human tumor specimens, the expression levels of USP39 and Cyclin B1 are positively relevant. CONCLUSION Our data support the evidence that USP39 acts a novel deubiquitinating enzyme of Cyclin B1 and promoted tumor cell proliferation at least in part through Cyclin B1 stabilization, represents a promising therapeutic strategy for tumor patients.
Collapse
Affiliation(s)
- Yue Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xinyi Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Weiwei Hu
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenjing Ma
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Haimei Tang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xibao Zhao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Guodong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Neurosurgery, Shenzhen Second People's Hospital, The first Affiliated Hospital of Shenzhen University, Shenzhen University Medical School, Shenzhen 518055, China; Institute of Biological Therapy, Shenzhen University, Shenzhen 518055, China.
| |
Collapse
|
8
|
Du X, Li S, Yang K, Cao Y. Downregulation of Sonic hedgehog signaling induces G2-arrest in genital warts. Skin Res Technol 2023; 29:e13265. [PMID: 36704875 PMCID: PMC9838784 DOI: 10.1111/srt.13265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Human papillomavirus (HPV) infected keratinocyte dysfunction results in the formation of genital warts, and the specific role of Sonic hedgehog (SHh) signaling in genital warts remains elusive. Thus, this study aimed to identify the correlation between wart formation and SHh signaling. MATERIALS AND METHODS In this study, nine male patients with genital warts were recruited, and the expression of SHh and its downstream signal molecules Patched-1 and GLI family zinc finger 1 (Ptch1 and Gli1) was detected. Moreover, G2-phase cells in the collected genital warts samples were assessed with normal foreskin samples as a comparison. HPV6/11 were detected via in situ hybridization (ISH), and SHh expression of the corresponding paraffin sections was determined via immunohistochemical staining (IHC). In addition, an in vitro down-regulated SHh model was constructed by siRNA transfection of the HaCaT cell line, and the cell cycle was detected at 36 h by flow cytometry with propidium iodide staining. RESULTS SHh, Ptch1, and Gli1 in warts were significantly downregulated in the condyloma acuminatum (CA) group compared to the normal foreskin group. G2-phase cells in the middle section of the spinous layer of CA wart tissues were significantly increased. Moreover, the expression of HPV-DNA was amplified and negatively correlated with SHh activity in CA wart tissues. Lastly, the downregulation of SHh-induced G2 arrest in vitro. CONCLUSIONS The downregulation of the SHh signaling promotes HPV replication and the formation of warts by inducing G2/M arrest in the keratinocytes of CA.
Collapse
Affiliation(s)
- Xiangxi Du
- Department of DermatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shan Li
- Department of AnaesthesiologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kun Yang
- Department of DermatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuchun Cao
- Department of DermatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
9
|
Fang H, Wang L, Yu L, Shen F, Yang Z, Yang Y, Li S, Dai H, Tan F, Lin J, Sheng H. Effects of metformin on Sonic hedgehog subgroup medulloblastoma progression: In vitro and in vivo studies. Front Pharmacol 2022; 13:928853. [PMID: 36278239 PMCID: PMC9585190 DOI: 10.3389/fphar.2022.928853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Metformin is a first-line drug for type 2 diabetes, and its anticancer effects have also been widely studied in recent years. The Sonic hedgehog (Shh) signaling pathway is involved in the initiation and progression of medulloblastoma. In order to develop a new treatment strategy for medulloblastoma (MB), this study investigated the inhibitory effect of metformin on MB and the underlying mechanism of metformin on the Shh signaling pathway. The effect of metformin on proliferation was evaluated by the cell counting kit-8 (CCK-8) test and colony formation experiment. The effect of metformin on metastasis was assessed by the scratch-wound assay and transwell invasion assay. Cell cycle and apoptosis were evaluated by flow cytometry, and the associated proteins were examined by western blotting. The mRNA and protein expression levels related to the Shh pathway were measured by quantitative PCR, western blotting, and immunofluorescence staining. The xenograft murine model was carried out to evaluate the anticancer effect of metformin on medulloblastoma in vivo. Metformin inhibited proliferation and metastasis of the Shh subgroup MB cell line, and the inhibitory effect on proliferation was related to apoptosis and the block of the cell cycle at the G0/G1 phase. Animal experiments showed that metformin inhibits medulloblastoma growth in vivo. Moreover, metformin decreased mRNA and protein expression levels of the Shh pathway, and this effect was reversed by the AMP-activated protein kinase (AMPK) siRNA. Furthermore, the pro-apoptotic and cell cycle arrest effects of metformin on Daoy cells could be reversed by the Shh pathway activators. Our findings demonstrated that metformin could inhibit medulloblastoma progression in vitro and in vivo, and this effect was associated with AMPK-mediated inhibition of the Shh signaling pathway in vitro studies.
Collapse
Affiliation(s)
- Huangyi Fang
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lingfei Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Lisheng Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fang Shen
- Department of Surgery, Box Hill Hospital Eastern Health, VIC, Australia
| | - Zelin Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shize Li
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haipeng Dai
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Feng Tan
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Feng Tan, ; Jian Lin, ; Hansong Sheng,
| | - Jian Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Feng Tan, ; Jian Lin, ; Hansong Sheng,
| | - Hansong Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Feng Tan, ; Jian Lin, ; Hansong Sheng,
| |
Collapse
|
10
|
Stagni F, Bartesaghi R. The Challenging Pathway of Treatment for Neurogenesis Impairment in Down Syndrome: Achievements and Perspectives. Front Cell Neurosci 2022; 16:903729. [PMID: 35634470 PMCID: PMC9130961 DOI: 10.3389/fncel.2022.903729] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS), also known as trisomy 21, is a genetic disorder caused by triplication of Chromosome 21. Gene triplication may compromise different body functions but invariably impairs intellectual abilities starting from infancy. Moreover, after the fourth decade of life people with DS are likely to develop Alzheimer’s disease. Neurogenesis impairment during fetal life stages and dendritic pathology emerging in early infancy are thought to be key determinants of alterations in brain functioning in DS. Although the progressive improvement in medical care has led to a notable increase in life expectancy for people with DS, there are currently no treatments for intellectual disability. Increasing evidence in mouse models of DS reveals that pharmacological interventions in the embryonic and neonatal periods may greatly benefit brain development and cognitive performance. The most striking results have been obtained with pharmacotherapies during embryonic life stages, indicating that it is possible to pharmacologically rescue the severe neurodevelopmental defects linked to the trisomic condition. These findings provide hope that similar benefits may be possible for people with DS. This review summarizes current knowledge regarding (i) the scope and timeline of neurogenesis (and dendritic) alterations in DS, in order to delineate suitable windows for treatment; (ii) the role of triplicated genes that are most likely to be the key determinants of these alterations, in order to highlight possible therapeutic targets; and (iii) prenatal and neonatal treatments that have proved to be effective in mouse models, in order to rationalize the choice of treatment for human application. Based on this body of evidence we will discuss prospects and challenges for fetal therapy in individuals with DS as a potential means of drastically counteracting the deleterious effects of gene triplication.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- *Correspondence: Renata Bartesaghi,
| |
Collapse
|
11
|
Wang W, Shiraishi R, Kawauchi D. Sonic Hedgehog Signaling in Cerebellar Development and Cancer. Front Cell Dev Biol 2022; 10:864035. [PMID: 35573667 PMCID: PMC9100414 DOI: 10.3389/fcell.2022.864035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
The sonic hedgehog (SHH) pathway regulates the development of the central nervous system in vertebrates. Aberrant regulation of SHH signaling pathways often causes neurodevelopmental diseases and brain tumors. In the cerebellum, SHH secreted by Purkinje cells is a potent mitogen for granule cell progenitors, which are the most abundant cell type in the mature brain. While a reduction in SHH signaling induces cerebellar structural abnormalities, such as hypoplasia in various genetic disorders, the constitutive activation of SHH signaling often induces medulloblastoma (MB), one of the most common pediatric malignant brain tumors. Based on the existing literature on canonical and non-canonical SHH signaling pathways, emerging basic and clinical studies are exploring novel therapeutic approaches for MB by targeting SHH signaling at distinct molecular levels. In this review, we discuss the present consensus on SHH signaling mechanisms, their roles in cerebellar development and tumorigenesis, and the recent advances in clinical trials for MB.
Collapse
Affiliation(s)
- Wanchen Wang
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ryo Shiraishi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- Department of NCNP Brain Physiology and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Kawauchi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- *Correspondence: Daisuke Kawauchi,
| |
Collapse
|
12
|
Kaushal JB, Batra SK, Rachagani S. Hedgehog signaling and its molecular perspective with cholesterol: a comprehensive review. Cell Mol Life Sci 2022; 79:266. [PMID: 35486193 PMCID: PMC9990174 DOI: 10.1007/s00018-022-04233-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/18/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Hedgehog (Hh) signaling is evolutionarily conserved and plays an instructional role in embryonic morphogenesis, organogenesis in various animals, and the central nervous system organization. Multiple feedback mechanisms dynamically regulate this pathway in a spatiotemporal and context-dependent manner to confer differential patterns in cell fate determination. Hh signaling is complex due to canonical and non-canonical mechanisms coordinating cell-cell communication. In addition, studies have demonstrated a regulatory framework of Hh signaling and shown that cholesterol is vital for Hh ligand biogenesis, signal generation, and transduction from the cell surface to intracellular space. Studies have shown the importance of a specific cholesterol pool, termed accessible cholesterol, which serves as a second messenger, conveying signals between smoothened (Smo) and patched 1 (Ptch1) across the plasma and ciliary membranes. Remarkably, recent high-resolution structural and molecular studies shed new light on the interplay between Hh signaling and cholesterol in membrane biology. These studies elucidated novel mechanistic insight into the release and dispersal of cholesterol-anchored Hh and the basis of Hh recognition by Ptch1. Additionally, the putative model of Smo activation by cholesterol binding and/or modification and Ptch1 antagonization of Smo has been explicated. However, the coupling mechanism of Hh signaling and cholesterol offered a new regulatory principle in cell biology: how effector molecules of the Hh signal network react to and remodel cholesterol accessibility in the membrane and selectively activate Hh signaling proteins thereof. Recognizing the biological importance of cholesterol in Hh signaling activation and transduction opens the door for translational research to develop novel therapeutic strategies. This review looks in-depth at canonical and non-canonical Hh signaling and the distinct proposed model of cholesterol-mediated regulation of Hh signaling components, facilitating a more sophisticated understanding of the Hh signal network and cholesterol biology.
Collapse
Affiliation(s)
- Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffet Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
13
|
Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. BIOLOGY 2022; 11:biology11040585. [PMID: 35453784 PMCID: PMC9027240 DOI: 10.3390/biology11040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and its incidence is steadily increasing. The development of HCC is a complex, multi-step process that is accompanied by alterations in multiple signaling cascades. Recent years have seen advancement in understanding molecular signaling pathways that play central roles in hepatocarcinogenesis. Aberrant activation of YAP/TAZ, Hedgehog, or Wnt/β-catenin signaling is frequently found in a subset of HCC patients. Targeting the signaling pathway via small molecule inhibitors could be a promising therapeutic option for the subset of patients. In this review, we will introduce the signaling pathways, discuss their roles in the development of HCC, and propose a therapeutic approach targeting the signaling pathways in the context of HCC. Abstract Hepatocellular carcinoma (HCC) is a major health concern worldwide, and its incidence is increasing steadily. To date, receptor tyrosine kinases (RTKs) are the most favored molecular targets for the treatment of HCC, followed by immune checkpoint regulators such as PD-1, PD-L1, and CTLA-4. With less than desirable clinical outcomes from RTK inhibitors as well as immune checkpoint inhibitors (ICI) so far, novel molecular target therapies have been proposed for HCC. In this review, we will introduce diverse molecular signaling pathways that are aberrantly activated in HCC, focusing on YAP/TAZ, Hedgehog, and Wnt/β-catenin signaling pathways, and discuss potential therapeutic strategies targeting the signaling pathways in HCC.
Collapse
|
14
|
Nguyen NM, Cho J. Hedgehog Pathway Inhibitors as Targeted Cancer Therapy and Strategies to Overcome Drug Resistance. Int J Mol Sci 2022; 23:ijms23031733. [PMID: 35163655 PMCID: PMC8835893 DOI: 10.3390/ijms23031733] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023] Open
Abstract
Hedgehog (Hh) signaling is a highly conserved pathway that plays a vital role during embryonic development. Recently, uncontrolled activation of this pathway has been demonstrated in various types of cancer. Therefore, Hh pathway inhibitors have emerged as an important class of anti-cancer agents. Unfortunately, however, their reputation has been tarnished by the emergence of resistance during therapy, necessitating clarification of mechanisms underlying the drug resistance. In this review, we briefly overview canonical and non-canonical Hh pathways and their inhibitors as targeted cancer therapy. In addition, we summarize the mechanisms of resistance to Smoothened (SMO) inhibitors, including point mutations of the drug binding pocket or downstream molecules of SMO, and non-canonical mechanisms to reinforce Hh pathway output. A distinct mechanism involving loss of primary cilia is also described to maintain GLI activity in resistant tumors. Finally, we address the main strategies to circumvent the drug resistance. These strategies include the development of novel and potent inhibitors targeting different components of the canonical Hh pathway or signaling molecules of the non-canonical pathway. Further studies are necessary to avoid emerging resistance to Hh inhibitors and establish an optimal customized regimen with improved therapeutic efficacy to treat various types of cancer, including basal cell carcinoma.
Collapse
|
15
|
Tao J, Chen Y, Zhuang Y, Wei R, Getachew A, Pan T, Yang F, Li Y. Inhibition of Hedgehog Delays Liver Regeneration through Disrupting the Cell Cycle. Curr Issues Mol Biol 2022; 44:470-482. [PMID: 35723318 PMCID: PMC8928988 DOI: 10.3390/cimb44020032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 11/16/2022] Open
Abstract
Liver regeneration is a complicated biological process orchestrated by various liver resident cells. Hepatic cell proliferation and reconstruction of the hepatic architecture involve multiple signaling pathways. It has been reported that the Hh signal is involved in liver regeneration. However, the signal transduction pathways and cell types involved are ill studied. This study aimed to investigate hedgehog signal response cell types and the specific molecular mechanism involved in the process of liver regeneration. Partial hepatectomy (PH) of 70% was performed on ICR (Institute of Cancer Research) mice to study the process of liver regeneration. We found that the hedgehog signal was activated significantly after PH, including hedgehog ligands, receptors and intracellular signaling molecules. Ligand signals were mainly expressed in bile duct cells and non-parenchymal hepatic cells, while receptors were expressed in hepatocytes and some non-parenchymal cells. Inhibition of the hedgehog signal treated with vismodegib reduced the liver regeneration rate after partial hepatectomy, including inhibition of hepatic cell proliferation by decreasing Cyclin D expression and disturbing the cell cycle through the accumulation of Cyclin B. The current study reveals the important role of the hedgehog signal and its participation in the regulation of hepatic cell proliferation and the cell cycle during liver regeneration. It provides new insight into the recovery of the liver after liver resection.
Collapse
Affiliation(s)
- Jiawang Tao
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Chen
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
| | - Yuanqi Zhuang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
| | - Ruzhi Wei
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
| | - Anteneh Getachew
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
| | - Tingcai Pan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
| | - Fan Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China;
| | - Yinxiong Li
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; (J.T.); (Y.C.); (Y.Z.); (R.W.); (A.G.); (T.P.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou 510530, China
- Correspondence: ; Tel.: +86-(020)-3201-5207
| |
Collapse
|
16
|
Polivka L, Parietti V, Bruneau J, Soucie E, Madrange M, Bayard E, Rignault R, Canioni D, Fraitag S, Lhermitte L, Feroul M, Tissandier M, Rossignol J, Frenzel L, Cagnard N, Meni C, Bouktit H, Collange AF, Gougoula C, Parisot M, Bader-Meunier B, Livideanu C, Laurent C, Arock M, Hadj-Rabia S, Rüther U, Dubreuil P, Bodemer C, Hermine O, Maouche-Chrétien L. The association of Greig syndrome and mastocytosis reveals the involvement of the hedgehog pathway in advanced mastocytosis. Blood 2021; 138:2396-2407. [PMID: 34424959 DOI: 10.1182/blood.2020010207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/28/2021] [Indexed: 11/20/2022] Open
Abstract
Mastocytosis is a heterogeneous disease characterized by an abnormal accumulation of mast cells (MCs) in 1 or several organs. Although a somatic KIT D816V mutation is detected in ∼85% of patients, attempts to demonstrate its oncogenic effect alone have repeatedly failed, suggesting that additional pathways are involved in MC transformation. From 3 children presenting with both Greig cephalopolysyndactyly syndrome (GCPS, Mendelian Inheritance in Man [175700]) and congenital mastocytosis, we demonstrated the involvement of the hedgehog (Hh) pathway in mastocytosis. GCPS is an extremely rare syndrome resulting from haploinsufficiency of GLI3, the major repressor of Hh family members. From these familial cases of mastocytosis, we demonstrate that the Hh pathway is barely active in normal primary MCs and is overactive in neoplastic MCs. GLI3 and KIT mutations had a synergistic, tumorigenic effect on the onset of mastocytosis in a GCPS mouse model. Finally, Hh inhibitors suppressed neoplastic MC proliferation in vitro and extend the survival time of mice with aggressive systemic mastocytosis (ASM). This work revealed, for the first time, the involvement of Hh signaling in the pathophysiology of mastocytosis and demonstrated the cooperative effects of the KIT and Hh oncogenic pathways in mice with ASM, leading to the identification of new promising therapeutic targets.
Collapse
Affiliation(s)
- L Polivka
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - V Parietti
- Department of Animal Experimentation, Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - J Bruneau
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - E Soucie
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France
| | - M Madrange
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - E Bayard
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - R Rignault
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - D Canioni
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - S Fraitag
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - L Lhermitte
- Institut Necker-Enfants Malades, Université de Paris, INSERM Unité (U)1151, Paris, France
- Laboratory of Onco-Hematology, Hôpital Universitaire Necker Enfants-Malades, AP-HP, Paris, France
| | - M Feroul
- Institut Necker-Enfants Malades, Université de Paris, INSERM Unité (U)1151, Paris, France
- Laboratory of Onco-Hematology, Hôpital Universitaire Necker Enfants-Malades, AP-HP, Paris, France
| | - M Tissandier
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - J Rossignol
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - L Frenzel
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Department of Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris-Centre University, Imagine Institute, Paris, France
| | - N Cagnard
- Bioinformatics, Platform Bioinformatics, INSERM U1163, Paris-Centre University, Imagine Institute, Paris, France
| | - C Meni
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - H Bouktit
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - A-F Collange
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - C Gougoula
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT), Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - M Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Service (UMS)3633, Paris-Centre University, Imagine Institute, Paris, France
| | - B Bader-Meunier
- Department of Pediatric Immunology and Hematology, Necker-Enfants Malades Hospital, AP-HP, INSERM U1163, Paris-Centre University, Paris, France
| | - C Livideanu
- Service de Dermatologie, CEREMAST, CHU de Toulouse
| | - C Laurent
- Service d'Anatomie-Pathologique, Oncopole, Centre Hospitalier de Universitaire (CJU) de Toulouse, Toulouse, France
| | - M Arock
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Centre de Recherche des Cordeliers, INSERM Unité de Recherche Mixte en Santé (UMRS)1138, Paris, France
- Laboratory of Hematology, Pitié-Salpêtrière Hospital, AP-HP Sorbonne Université, Paris, France; and
| | - S Hadj-Rabia
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - U Rüther
- Institute of Animal Developmental and Molecular Biology, Heinrich Heine University, Düsseldorf, Germany
| | - P Dubreuil
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France
| | - C Bodemer
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - O Hermine
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Department of Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris-Centre University, Imagine Institute, Paris, France
| | - L Maouche-Chrétien
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| |
Collapse
|
17
|
Tarulli GA, Cripps SM, Pask AJ, Renfree MB. Spatiotemporal map of key signaling factors during early penis development. Dev Dyn 2021; 251:609-624. [PMID: 34697862 PMCID: PMC9539974 DOI: 10.1002/dvdy.433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/27/2021] [Accepted: 09/28/2021] [Indexed: 12/31/2022] Open
Abstract
The formation of the external genitalia is a highly complex developmental process, considering it involves a wide range of cell types and results in sexually dimorphic outcomes. Development is controlled by several secreted signalling factors produced in complex spatiotemporal patterns, including the hedgehog (HH), bone morphogenic protein (BMP), fibroblast growth factor (FGF) and WNT signalling families. Many of these factors act on or are influenced by the actions of the androgen receptor (AR) that is critical to masculinisation. This complexity of expression makes it difficult to conceptualise patterns of potential importance. Mapping expression during key stages of development is needed to develop a comprehensive model of how different cell types interact in formation of external genitalia, and the global regulatory networks at play. This is particularly true in light of the sensitivity of this process to environmental disruption during key stages of development. The goal of this review is to integrate all recent studies on gene expression in early penis development to create a comprehensive spatiotemporal map. This serves as a resource to aid in visualising potentially significant interactions involved in external genital development.
Diagrams of published RNA and protein localisation data for key secreted signalling factors during early penis development. Unconventional expression patterns are identified that suggest novel signalling axes during development. Key research gaps and limitations are identified and discussed.
Collapse
Affiliation(s)
- Gerard A Tarulli
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Samuel M Cripps
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Hedgehog/GLI Signaling Pathway: Transduction, Regulation, and Implications for Disease. Cancers (Basel) 2021; 13:cancers13143410. [PMID: 34298625 PMCID: PMC8304605 DOI: 10.3390/cancers13143410] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The Hedgehog/GLI (Hh/GLI) pathway plays a major role during development and it is commonly dysregulated in many diseases, including cancer. This highly concerted series of ligands, receptors, cytoplasmic signaling molecules, transcription factors, and co-regulators is involved in regulating the biological functions controlled by this pathway. Activation of Hh/GLI in cancer is most often through a non-canonical method of activation, independent of ligand binding. This review is intended to summarize our current understanding of the Hh/GLI signaling, non-canonical mechanisms of pathway activation, its implication in disease, and the current therapeutic strategies targeting this cascade. Abstract The Hh/GLI signaling pathway was originally discovered in Drosophila as a major regulator of segment patterning in development. This pathway consists of a series of ligands (Shh, Ihh, and Dhh), transmembrane receptors (Ptch1 and Ptch2), transcription factors (GLI1–3), and signaling regulators (SMO, HHIP, SUFU, PKA, CK1, GSK3β, etc.) that work in concert to repress (Ptch1, Ptch2, SUFU, PKA, CK1, GSK3β) or activate (Shh, Ihh, Dhh, SMO, GLI1–3) the signaling cascade. Not long after the initial discovery, dysregulation of the Hh/GLI signaling pathway was implicated in human disease. Activation of this signaling pathway is observed in many types of cancer, including basal cell carcinoma, medulloblastoma, colorectal, prostate, pancreatic, and many more. Most often, the activation of the Hh/GLI pathway in cancer occurs through a ligand-independent mechanism. However, in benign disease, this activation is mostly ligand-dependent. The upstream signaling component of the receptor complex, SMO, is bypassed, and the GLI family of transcription factors can be activated regardless of ligand binding. Additional mechanisms of pathway activation exist whereby the entirety of the downstream signaling pathway is bypassed, and PTCH1 promotes cell cycle progression and prevents caspase-mediated apoptosis. Throughout this review, we summarize each component of the signaling cascade, non-canonical modes of pathway activation, and the implications in human disease, including cancer.
Collapse
|
19
|
Kotulak-Chrząszcz A, Kmieć Z, Wierzbicki PM. Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review). Int J Mol Med 2021; 47:106. [PMID: 33907821 PMCID: PMC8057295 DOI: 10.3892/ijmm.2021.4939] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/10/2021] [Indexed: 01/07/2023] Open
Abstract
Cancers of the urinary tract, as well as those of the female and male reproductive systems, account for a large percentage of malignancies worldwide. Mortality is frequently affected by late diagnosis or therapeutic difficulties. The Sonic Hedgehog (SHH) pathway is an evolutionary conserved molecular cascade, which is mainly associated with the development of the central nervous system in fetal life. The present review aimed to provide an in‑depth summary of the SHH signaling pathway, including the characterization of its major components, the mechanism of its upstream regulation and non‑canonical activation, as well as its interactions with other cellular pathways. In addition, the three possible mechanisms of the cellular SHH cascade in cancer tissue are discussed. The aim of the present review was to summarize significant findings with regards to the expression of the SHH pathway components in kidney, bladder, ovarian, cervical and prostate cancer. Reports associated with common deficits and de‑regulations of the SHH pathway were summarized, despite the differences in molecular and histological patterns among these malignancies. However, currently, neither are SHH pathway elements included in panels of prognostic/therapeutic molecular patterns in any of the discussed cancers, nor have the drugs targeting SMO or GLIs been approved for therapy. The findings of the present review may support future studies on the treatment of and/or molecular targets for gynecological and genitourinary cancers.
Collapse
Affiliation(s)
| | | | - Piotr M. Wierzbicki
- Correspondence to: Dr Piotr M. Wierzbicki, Department of Histology, Faculty of Medicine, Medical University of Gdansk, ul. Debinki 1, 80211 Gdansk, Poland, E-mail:
| |
Collapse
|
20
|
Yamashita K, Tamura S, Honsho M, Yada H, Yagita Y, Kosako H, Fujiki Y. Mitotic phosphorylation of Pex14p regulates peroxisomal import machinery. J Cell Biol 2021; 219:152047. [PMID: 32854114 PMCID: PMC7659713 DOI: 10.1083/jcb.202001003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/28/2020] [Accepted: 07/13/2020] [Indexed: 01/27/2023] Open
Abstract
Peroxisomal matrix proteins are imported into peroxisomes via membrane-bound docking/translocation machinery. One central component of this machinery is Pex14p, a peroxisomal membrane protein involved in the docking of Pex5p, the receptor for peroxisome targeting signal type 1 (PTS1). Studies in several yeast species have shown that Pex14p is phosphorylated in vivo, whereas no function has been assigned to Pex14p phosphorylation in yeast and mammalian cells. Here, we investigated peroxisomal protein import and its dynamics in mitotic mammalian cells. In mitotically arrested cells, Pex14p is phosphorylated at Ser-232, resulting in a lower import efficiency of catalase, but not the majority of proteins including canonical PTS1 proteins. Conformational change induced by the mitotic phosphorylation of Pex14p more likely increases homomeric interacting affinity and suppresses topological change of its N-terminal part, thereby giving rise to the retardation of Pex5p export in mitotic cells. Taken together, these data show that mitotic phosphorylation of Pex14p and consequent suppression of catalase import are a mechanism of protecting DNA upon nuclear envelope breakdown at mitosis.
Collapse
Affiliation(s)
- Koichiro Yamashita
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | | | - Masanori Honsho
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Institute of Rheological Functions of Food, Fukuoka, Japan
| | - Hiroto Yada
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Yagita
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Yukio Fujiki
- Division of Organelle Homeostasis, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Institute of Rheological Functions of Food, Fukuoka, Japan
| |
Collapse
|
21
|
Jiang Z, Derrick-Roberts AL, Byers S. Altered IHH signaling contributes to reduced chondrocyte proliferation in the growth plate of MPS VII mice. Mol Genet Metab Rep 2020; 25:100668. [PMID: 33117654 PMCID: PMC7582094 DOI: 10.1016/j.ymgmr.2020.100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 10/29/2022] Open
Abstract
Bone elongation is driven by chondrocyte proliferation and hypertrophy in the growth plate. Both processes are modulated by multiple signaling pathways including the Indian Hedgehog (IHH) signaling pathway. Mucopolysaccharidoses (MPS) are a group of lysosomal storage disorders characterized by accumulation of glycosaminoglycans (GAGs) in multiple tissues and organs, leading to a range of clinical symptoms including bone shortening through mechanisms that are not fully understood. Using MPS VII mice, we previously observed a reduction in the number of proliferating and hypertrophic chondrocytes and a reduced gene expression of Ihh in the tibial growth plate. We further demonstrate here that IHH secretion by MPS VII chondrocytes was reduced both in vitro and in vivo. While normal chondrocytes showed no response to exogenous IHH, proliferation of MPS VII chondrocytes was stimulated in response to exogenous IHH in vitro. This was accompanied by an elevated gene expression of patched receptor (Ptch1). The results from this study suggested that reduced proliferation in MPS VII growth plate may be partially due to dysfunction of the IHH signaling pathway.
Collapse
Affiliation(s)
- Zhirui Jiang
- Genetics and Evolution, The University of Adelaide, Adelaide, SA, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Ainslie L.K. Derrick-Roberts
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Paediatrics, The University of Adelaide, Adelaide, SA, Australia
| | - Sharon Byers
- Genetics and Evolution, The University of Adelaide, Adelaide, SA, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Paediatrics, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
22
|
Onodera S, Nakamura Y, Azuma T. Gorlin Syndrome: Recent Advances in Genetic Testing and Molecular and Cellular Biological Research. Int J Mol Sci 2020; 21:E7559. [PMID: 33066274 PMCID: PMC7590212 DOI: 10.3390/ijms21207559] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023] Open
Abstract
Gorlin syndrome is a skeletal disorder caused by a gain of function mutation in Hedgehog (Hh) signaling. The Hh family comprises of many signaling mediators, which, through complex mechanisms, play several important roles in various stages of development. The Hh information pathway is essential for bone tissue development. It is also the major driver gene in the development of basal cell carcinoma and medulloblastoma. In this review, we first present the recent advances in Gorlin syndrome research, in particular, the signaling mediators of the Hh pathway and their functions at the genetic level. Then, we discuss the phenotypes of mutant mice and Hh signaling-related molecules in humans revealed by studies using induced pluripotent stem cells.
Collapse
Affiliation(s)
- Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kandamisaki-cho Chiyoda-ku, Tokyo 101-0061, Japan;
| | - Yuriko Nakamura
- Department of Oral Oncology, Oral and Maxillofacial Surgery, Tokyo Dental College, 5-11-13 Sugano, Ichikawa, Chiba 272-8513, Japan;
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kandamisaki-cho Chiyoda-ku, Tokyo 101-0061, Japan;
| |
Collapse
|
23
|
Doheny D, Manore SG, Wong GL, Lo HW. Hedgehog Signaling and Truncated GLI1 in Cancer. Cells 2020; 9:cells9092114. [PMID: 32957513 PMCID: PMC7565963 DOI: 10.3390/cells9092114] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
The hedgehog (HH) signaling pathway regulates normal cell growth and differentiation. As a consequence of improper control, aberrant HH signaling results in tumorigenesis and supports aggressive phenotypes of human cancers, such as neoplastic transformation, tumor progression, metastasis, and drug resistance. Canonical activation of HH signaling occurs through binding of HH ligands to the transmembrane receptor Patched 1 (PTCH1), which derepresses the transmembrane G protein-coupled receptor Smoothened (SMO). Consequently, the glioma-associated oncogene homolog 1 (GLI1) zinc-finger transcription factors, the terminal effectors of the HH pathway, are released from suppressor of fused (SUFU)-mediated cytoplasmic sequestration, permitting nuclear translocation and activation of target genes. Aberrant activation of this pathway has been implicated in several cancer types, including medulloblastoma, rhabdomyosarcoma, basal cell carcinoma, glioblastoma, and cancers of lung, colon, stomach, pancreas, ovarian, and breast. Therefore, several components of the HH pathway are under investigation for targeted cancer therapy, particularly GLI1 and SMO. GLI1 transcripts are reported to undergo alternative splicing to produce truncated variants: loss-of-function GLI1ΔN and gain-of-function truncated GLI1 (tGLI1). This review covers the biochemical steps necessary for propagation of the HH activating signal and the involvement of aberrant HH signaling in human cancers, with a highlight on the tumor-specific gain-of-function tGLI1 isoform.
Collapse
Affiliation(s)
- Daniel Doheny
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.D.); (S.G.M.); (G.L.W.)
| | - Sara G. Manore
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.D.); (S.G.M.); (G.L.W.)
| | - Grace L. Wong
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.D.); (S.G.M.); (G.L.W.)
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.D.); (S.G.M.); (G.L.W.)
- Wake Forest Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
- Correspondence: ; Tel.: +1-336-716-0695
| |
Collapse
|
24
|
Grund-Gröschke S, Ortner D, Szenes-Nagy AB, Zaborsky N, Weiss R, Neureiter D, Wipplinger M, Risch A, Hammerl P, Greil R, Sibilia M, Gratz IK, Stoitzner P, Aberger F. Epidermal activation of Hedgehog signaling establishes an immunosuppressive microenvironment in basal cell carcinoma by modulating skin immunity. Mol Oncol 2020; 14:1930-1946. [PMID: 32615027 PMCID: PMC7463314 DOI: 10.1002/1878-0261.12758] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/27/2020] [Accepted: 06/26/2020] [Indexed: 12/24/2022] Open
Abstract
Genetic activation of hedgehog/glioma‐associated oncogene homolog (HH/GLI) signaling causes basal cell carcinoma (BCC), a very frequent nonmelanoma skin cancer. Small molecule targeting of the essential HH effector Smoothened (SMO) has proven an effective therapy of BCC, though the frequent development of drug resistance poses major challenges to anti‐HH treatments. In light of recent breakthroughs in cancer immunotherapy, we analyzed the possible immunosuppressive mechanisms in HH/GLI‐induced BCC in detail. Using a genetic mouse model of BCC, we identified profound differences in the infiltration of BCC lesions with cells of the adaptive and innate immune system. Epidermal activation of Hh/Gli signaling led to an accumulation of immunosuppressive regulatory T cells, and to an increased expression of immune checkpoint molecules including programmed death (PD)‐1/PD‐ligand 1. Anti‐PD‐1 monotherapy, however, did not reduce tumor growth, presumably due to the lack of immunogenic mutations in common BCC mouse models, as shown by whole‐exome sequencing. BCC lesions also displayed a marked infiltration with neutrophils, the depletion of which unexpectedly promoted BCC growth. The study provides a comprehensive survey of and novel insights into the immune status of murine BCC and serves as a basis for the design of efficacious rational combination treatments. This study also underlines the need for predictive immunogenic mouse models of BCC to evaluate the efficacy of immunotherapeutic strategies in vivo.
Collapse
Affiliation(s)
- Sandra Grund-Gröschke
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Daniela Ortner
- Department of Dermatology, Venereology & Allergology, Medical University Innsbruck, Austria
| | - Antal B Szenes-Nagy
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Nadja Zaborsky
- IIIrd Medical Department, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg, Cancer Cluster Salzburg, Austria
| | - Richard Weiss
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University Salzburg, Cancer Cluster Salzburg, Austria
| | - Martin Wipplinger
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Angela Risch
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Peter Hammerl
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg, Cancer Cluster Salzburg, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Austria
| | - Iris K Gratz
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & Allergology, Medical University Innsbruck, Austria
| | - Fritz Aberger
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, Austria
| |
Collapse
|
25
|
Zhong W, Zhao H, Huang W, Zhang M, Zhang Q, Zhang Y, Chen C, Nueraihemaiti Z, Tuerhong D, Huang H, Maimaitili G, Chen F, Lin J. Identification of rare PTCH1 nonsense variant causing orofacial cleft in a Chinese family and an up-to-date genotype-phenotype analysis. Genes Dis 2020; 8:689-697. [PMID: 34291140 PMCID: PMC8278535 DOI: 10.1016/j.gendis.2019.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/10/2019] [Accepted: 12/31/2019] [Indexed: 01/09/2023] Open
Abstract
The Patched 1 (PTCH1) gene encodes a membrane receptor involved in the Hedgehog (Hh) signaling pathway, an abnormal state of which may result in congenital defects or human tumors. In this study, we conducted whole-exome sequencing on a three-generation Chinese family characterized with variable penetrance of orofacial clefts. A rare heterozygous variant in the PTCH1 gene (c.2833C > T p.R945X) was identified as a disease-associated mutation. Structural modeling revealed a truncation starting from the middle of the second extracellular domain of PTCH1 protein. This may damage its ligand recognition and sterol transportation abilities, thereby affecting the Hh signaling pathway. Biochemical assays indicated that the R945X protein had reduced stability compared to the wild-type in vitro. In addition, we reviewed the locations and mutation types of PTCH1 variants in individuals with clefting phenotypes, and analyzed the associations between clefts and locations or types of variants within PTCH1. Our findings provide further evidence that PTCH1 variants result in orofacial clefts, and contributed to genetic counseling and clinical surveillance in this family.
Collapse
Affiliation(s)
- Wenjie Zhong
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Huaxiang Zhao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Wenbin Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Mengqi Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Qian Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yue Zhang
- Department of Stomatology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, PR China
| | - Chong Chen
- Department of Stomatology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, PR China
| | | | | | - Huizhe Huang
- Chongqing Medical University, Chongqing, 400016, PR China
| | - Gulibaha Maimaitili
- Department of Stomatology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, PR China
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Jiuxiang Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| |
Collapse
|
26
|
Penela P, Ribas C, Sánchez-Madrid F, Mayor F. G protein-coupled receptor kinase 2 (GRK2) as a multifunctional signaling hub. Cell Mol Life Sci 2019; 76:4423-4446. [PMID: 31432234 PMCID: PMC6841920 DOI: 10.1007/s00018-019-03274-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates that G protein-coupled receptor kinase 2 (GRK2) is a versatile protein that acts as a signaling hub by modulating G protein-coupled receptor (GPCR) signaling and also via phosphorylation or scaffolding interactions with an extensive number of non-GPCR cellular partners. GRK2 multifunctionality arises from its multidomain structure and from complex mechanisms of regulation of its expression levels, activity, and localization within the cell, what allows the precise spatio-temporal shaping of GRK2 targets. A better understanding of the GRK2 interactome and its modulation mechanisms is helping to identify the GRK2-interacting proteins and its substrates involved in the participation of this kinase in different cellular processes and pathophysiological contexts.
Collapse
Affiliation(s)
- Petronila Penela
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Catalina Ribas
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
- Cell-Cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain.
| |
Collapse
|
27
|
Iulianella A, Stanton-Turcotte D. The Hedgehog receptor Patched1 regulates proliferation, neurogenesis, and axon guidance in the embryonic spinal cord. Mech Dev 2019; 160:103577. [PMID: 31634536 DOI: 10.1016/j.mod.2019.103577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/03/2019] [Accepted: 10/02/2019] [Indexed: 12/31/2022]
Abstract
The formation of the vertebrate nervous system depends on the complex interplay of morphogen signaling pathways and cell cycle progression to establish distinct cell fates. The Sonic hedgehog (Shh) signaling pathway is well understood to promote ventral cell fates in the developing spinal cord. A key regulator of Shh signaling is its receptor Patched1 (Ptch1). However, because the Ptch1 null mutation is lethal early in mouse embryogenesis, its role in controlling cell cycle progression, neurogenesis, and axon guidance in the developing spinal cord is not fully understood. An allele of Ptch1 called Wiggable (Ptch1Wig), which was previously shown to enhance Shh signaling, was used to test its ability to regulate neurogenesis and proliferation in the developing spinal cord. Ptch1Wig/Wig mutants displayed enhanced ventral proneural gene activation, and aberrant proliferation of the neural tube and floor plate cells, the latter normally being a quiescent population. The expression of the cell cycle regulators p27Kip1 and p57Kip2 were expanded in Ptch1Wig/Wig mutant spinal cords, as was the number of mitotic and S-phase nuclei, suggesting enhanced cell cycle progression. However, Ptch1Wig/Wig mutants also showed enhanced apoptosis in the ventral embryonic spinal cord, which resulted in thinner spinal cords at later embryonic stages. Commissural axons largely failed to cross the floor plate of Ptch1Wig/Wig mutant embryos, suggesting enhanced Shh signaling in these mutants led to a dorsal expansion of the chemoattraction front. These findings are consistent with a role of Ptch1 in regulating neurogenesis and proliferation of neural progenitors, and in restricting the influence of Shh signaling in commissural axon guidance to the floor plate.
Collapse
Affiliation(s)
- Angelo Iulianella
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Centre, Dalhousie University, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada.
| | - Danielle Stanton-Turcotte
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Centre, Dalhousie University, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia B3H-4R2, Canada
| |
Collapse
|
28
|
Niyaz M, Khan MS, Mudassar S. Hedgehog Signaling: An Achilles' Heel in Cancer. Transl Oncol 2019; 12:1334-1344. [PMID: 31352196 PMCID: PMC6664200 DOI: 10.1016/j.tranon.2019.07.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
Hedgehog signaling pathway originally identified in the fruit fly Drosophila is an evolutionarily conserved signaling mechanism with crucial roles in embryogenesis, growth and patterning. It exerts its biological effect through a signaling mechanism that terminates at glioma-associated oncogene (GLI) transcription factors which alternate between activator and repressor forms and mediate various responses. The important components of the pathway include the hedgehog ligands (SHH), the Patched (PTCH) receptor, Smoothened (SMO), Suppressor of Fused (SuFu) and GLI transcription factors. Activating or inactivating mutations in key genes cause uncontrolled activation of the pathway in a ligand independent manner. The ligand-dependent aberrant activation of the hedgehog pathway causing overexpression of hedgehog pathway components and its target genes occurs in autocrine as well as paracrine fashion. In adults, aberrant activation of hedgehog signaling has been linked to birth defects and multiple solid cancers. In this review, we assimilate data from recent studies to understand the mechanism of functioning of the hedgehog signaling pathway, role in cancer, its association in various solid malignancies and the current strategies being used to target this pathway for cancer treatment.
Collapse
Affiliation(s)
- Madiha Niyaz
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Soura, - 190011 Srinagar, Kashmir
| | - Mosin S Khan
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Soura, - 190011 Srinagar, Kashmir
| | - Syed Mudassar
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Soura, - 190011 Srinagar, Kashmir.
| |
Collapse
|
29
|
Pathania AS, Ren X, Mahdi MY, Shackleford GM, Erdreich-Epstein A. GRK2 promotes growth of medulloblastoma cells and protects them from chemotherapy-induced apoptosis. Sci Rep 2019; 9:13902. [PMID: 31554835 PMCID: PMC6761358 DOI: 10.1038/s41598-019-50157-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
G-protein coupled receptor kinase 2 (GRK2; ADRBK1, BARK1) is most known as a regulator of G-protein coupled receptors. However, GRK2 also has other functions. Medulloblastomas are the most common malignant brain cancers in children. GRK2 has not been implicated in medulloblastoma biology. Here we report that GRK2 knockdown slowed cell growth, diminished proliferation, and enhanced cisplatin- and etoposide-induced apoptosis in medulloblastoma cell lines UW228-2 and Daoy. Reciprocally, GRK2 overexpression attenuated apoptosis induced by these chemotherapy drugs. Cisplatin and etoposide increased phosphorylation of AKT (S473) and GRK2 knockdown mitigated this increase. Cisplatin and etoposide attenuated ERK phosphorylation, but GRK2 knockdown did not alter this effect. Wildtype GRK2 reversed the increase in cisplatin- and etoposide-induced apoptosis caused by GRK2 knockdown. GRK2-K220R (kinase dead) and GRK2-S670A (unphosphorylated, constitutively active) conferred protection from cisplatin that was similar to wildtype GRK2, suggesting that this protection may be mediated though a kinase-independent activity of GRK2. These data demonstrate that GRK2 contributes to proliferation and survival of these medulloblastoma cell lines and to their protection from cisplatin- and etoposide-induced apoptosis.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xiuhai Ren
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Min Y Mahdi
- Department of Radiology, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Gregory M Shackleford
- Department of Radiology, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Anat Erdreich-Epstein
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles and Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA.
- Department of Pathology, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
30
|
Zheng F, Xiao X, Wang C. Retracted: The Effect of PTCH1 on Ovarian Cancer Cell Proliferation and Apoptosis. Cancer Biother Radiopharm 2019; 34:103-109. [DOI: 10.1089/cbr.2018.2626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Fang Zheng
- Department of Gynaecology, Huangshi Aikang Hospital, Huangshi, China
| | - Xinyi Xiao
- Department of Gynaecology, Huangshi Aikang Hospital, Huangshi, China
| | - Chunmei Wang
- Department of Gynaecology, Huangshi Aikang Hospital, Huangshi, China
| |
Collapse
|
31
|
Fleet AJ, Hamel PA. The protein-specific activities of the transmembrane modules of Ptch1 and Ptch2 are determined by their adjacent protein domains. J Biol Chem 2018; 293:16583-16595. [PMID: 30166346 PMCID: PMC6204896 DOI: 10.1074/jbc.ra118.004478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/17/2018] [Indexed: 01/20/2023] Open
Abstract
Signaling through the Hedgehog (Hh) pathway is mediated by the Patched (Ptch) family of proteins. Although the vertebrate Ptch proteins Ptch1 and Ptch2 harbor two closely related transmembrane modules related to sterol-sensing domains (SSDs), the role of these closely related receptors in the Hh pathway are not equivalent. Ptch1 is essential for development and appears to be the principal receptor mediating responses to Hh ligands, whereas Ptch2 is nonessential, and its role in Hh-signaling remains ambiguous. We hypothesized that the SSDs of the Ptch proteins function as generic modules whose protein-specific activities are determined by the adjacent cytoplasmic and luminal domains. We first showed that individual N-terminal and C-terminal halves of Ptch1 associated noncovalently to mediate ligand-dependent regulation of Hh signaling. The analogous regions of Ptch2 also interacted noncovalently but did not repress the Hh pathway. However, the SSD of Ptch2 were capable of repressing Hh signaling, as determined using chimeric proteins where the SSDs of Ptch1 were replaced by those from Ptch2. Replacement of the SSDs of Ptch1 with the analogous regions from the cholesterol transporter NPC1 failed to produce a chimeric protein capable of Hh repression. Further refinement of the specific regions in Ptch1 and Ptch2 revealed that specific cytoplasmic domains of Ptch1 were necessary but not sufficient for repression of Hh signaling and that the two principal luminal domains of Ptch1 and Ptch2 were interchangeable. These data support a model where the SSDs of the Ptch family proteins exhibit generic activities and that the adjacent cytoplasmic and luminal domains determine their protein-specific activities.
Collapse
Affiliation(s)
- Andrew J Fleet
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Paul A Hamel
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
32
|
Dai J, Lin K, Huang Y, Lu Y, Chen WQ, Zhang XR, He BS, Pan YQ, Wang SK, Fan WX. Identification of critically carcinogenesis-related genes in basal cell carcinoma. Onco Targets Ther 2018; 11:6957-6967. [PMID: 30410353 PMCID: PMC6199216 DOI: 10.2147/ott.s170504] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Basal cell carcinoma (BCC) is a frequent malignant tumor of skin cancers with high morbidity. The objective of this study was to identify critical genes and pathways related to the carcinogenesis of BCC and gain more insights into the underlying molecular mechanisms of BCC. Materials and methods The gene expression profiles of GSE7553 and GSE103439 were downloaded from the Gene Expression Omnibus database with 19 tumors and 6 normal skin tissues. Differentially expressed genes (DEGs) were screened between BCC samples and normal tissues, followed by gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. Subsequently, protein–protein interaction (PPI) network was constructed for these DEGs, and module analysis was performed. Results A total of 313 DEGs were obtained. Among them, 222 genes were upregulated and 91 genes were downregulated. Enrichment analysis indicated that the upregulated genes were significantly enriched in cell cycle and mitosis, while the downregulated genes were mainly associated with unsaturated fatty acid metabolic process and cell differentiation. In addition, TOP2A, CDK1, and CCNB1 were identified as the top three hub genes ranked by degrees in the PPI network. Meanwhile, three subnetworks were derived, which indicated that these DEGs were significantly enriched in pathways, including “cell cycle”, “extracellular matrix–receptor interaction”, “basal cell carcinoma”, and “hedgehog signaling pathway”. Conclusions The novel critical DEGs and pathways identified in this study may serve pivotal roles in the carcinogenesis of BCC and indicate more molecular targets for the treatment of BCC.
Collapse
Affiliation(s)
- Jie Dai
- Department of Dermatology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kang Lin
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Yan Huang
- Department of Ultrasound, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Lu
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China,
| | - Wen-Qi Chen
- Department of Dermatology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Rong Zhang
- Department of Dermatology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bang-Shun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Qin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shu-Kui Wang
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China,
| | - Wei-Xin Fan
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China,
| |
Collapse
|
33
|
Non-Canonical Hedgehog Signaling Is a Positive Regulator of the WNT Pathway and Is Required for the Survival of Colon Cancer Stem Cells. Cell Rep 2018; 21:2813-2828. [PMID: 29212028 DOI: 10.1016/j.celrep.2017.11.025] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/15/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022] Open
Abstract
Colon cancer is a heterogeneous tumor driven by a subpopulation of cancer stem cells (CSCs). To study CSCs in colon cancer, we used limiting dilution spheroid and serial xenotransplantation assays to functionally define the frequency of CSCs in a panel of patient-derived cancer organoids. These studies demonstrated cancer organoids to be enriched for CSCs, which varied in frequency between tumors. Whole-transcriptome analysis identified WNT and Hedgehog signaling components to be enhanced in CSC-enriched tumors and in aldehyde dehydrogenase (ALDH)-positive CSCs. Canonical GLI-dependent Hedgehog signaling is a negative regulator of WNT signaling in normal intestine and intestinal tumors. Here, we show that Hedgehog signaling in colon CSCs is autocrine SHH-dependent, non-canonical PTCH1 dependent, and GLI independent. In addition, using small-molecule inhibitors and RNAi against SHH-palmitoylating Hedgehog acyltransferase (HHAT), we demonstrate that non-canonical Hedgehog signaling is a positive regulator of WNT signaling and required for colon CSC survival.
Collapse
|
34
|
Id1 and Sonic Hedgehog Mediate Cell Cycle Reentry and Apoptosis Induced by Amyloid Beta-Peptide in Post-mitotic Cortical Neurons. Mol Neurobiol 2018; 56:465-489. [PMID: 29721855 DOI: 10.1007/s12035-018-1098-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/24/2018] [Indexed: 10/17/2022]
Abstract
Amyloid beta-peptide (Aβ), the neurotoxic component of senile plaques in Alzheimer's disease (AD) brains, is known to trigger cell cycle reentry in post-mitotic neurons followed by apoptosis. However, the underlying mechanisms remain unclear. Recently, we have reported that Aβs stimulate the expression of inhibitor of differentiation-1 (Id1) to induce sonic hedgehog (SHH) (Hung et al., Mol Neurobiol 53(2):793-809, 2016), and both are mitogens capable of triggering cell cycle progression. In this work, we tested the hypothesis that Aβ-induced Id1 and SHH contribute to cell cycle reentry leading to apoptosis in neurons. We found that Aβ triggered cell cycle progression in the post-mitotic neurons, as indicated by the increased expression of two G1-phase markers including cyclin D1 and phosphorylated retinoblastoma protein (pRb), two G2-phase markers such as proliferating cell nuclear antigen (PCNA) and incorporation of 5-bromo-2'-deoxyuridine (BrdU) into newly synthesized DNA, as well as the mitotic marker histone H3 phosphorylated at Ser-10. As expected, Aβ also enhanced caspase-3 cleavage in the cortical neurons. Id1 siRNA, the neutralization antibody against SHH (SHH-Ab), and the cyclin-dependent kinase (CDK)-4/6 inhibitor PD0332991 all attenuated, in part or in full, the Aβ-induced expression of these cell cycle markers. Indeed, exogenous recombinant Id1 protein and the biologically active N-terminal fragment of SHH (SHH-N) were both sufficient to enhance the expression of cell cycle markers independent of Aβ. Taken together, our results revealed the critical roles of Id1 and SHH mediating Aβ-dependent cell cycle reentry and subsequently caspase-dependent apoptosis in the fully differentiated post-mitotic neurons, at least in vitro.
Collapse
|
35
|
Abstract
Ever since its initial discovery in Drosophila, hedgehog signaling has been linked to foregut development, The mammalian genome expresses three Hedgehog paralogues, sonic hedgehog (Shh), Indian Hedgehog, and desert hedgehog. In the mucosa of the embryonic and adult foregut, Shh expression is the highest. It has now become clear that hedgehog signaling is of pivotal importance in gastric homeostasis. Aberrant activation of hedgehog signaling is associated with a range of pathological consequences including various cancers. Also in gastric cancer, clinical and preclinical data support a role of Hedgehog signaling in neoplastic transformation, and gastrointestinal cancer development, also through cancer stroma interaction. Technological advance are facilitating monitoring Hedgehog signaling broadening options for the more efficient screening of individuals predisposed to eventually developing gastric cancer and targeting Hedgehog signaling may provide opportunities for prophylactic therapy once atrophic gastritis develops. Nevertheless, convincing evidence that Hedgehog antagonists are of clinically useful in the context of gastric cancer is still conspicuously lacking. Here we analyze review the role of Hedgehog in gastric physiology and the potential usefulness of targeting Hedgehog signaling in gastric cancer.
Collapse
Affiliation(s)
- Adamu Ishaku Akyala
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University, Rotterdam, Rotterdam, The Netherlands.,Department of Microbiology, Faculty of Natural and Applied Sciences Nasarawa State University, Keffi, Nasarawa, Nigeria
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University, Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
36
|
Verdelho Machado M, Diehl AM. The hedgehog pathway in nonalcoholic fatty liver disease. Crit Rev Biochem Mol Biol 2018; 53:264-278. [PMID: 29557675 DOI: 10.1080/10409238.2018.1448752] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of obesity-associated liver diseases and it has become the major cause of cirrhosis in the Western world. The high prevalence of NAFLD-associated advanced liver disease reflects both the high prevalence of obesity-related fatty liver (hepatic steatosis) and the lack of specific treatments to prevent hepatic steatosis from progressing to more serious forms of liver damage, including nonalcoholic steatohepatitis (NASH), cirrhosis, and primary liver cancer. The pathogenesis of NAFLD is complex, and not fully understood. However, compelling evidence demonstrates that dysregulation of the hedgehog (Hh) pathway is involved in both the pathogenesis of hepatic steatosis and the progression from hepatic steatosis to more serious forms of liver damage. Inhibiting hedgehog signaling enhances hepatic steatosis, a condition which seldom results in liver-related morbidity or mortality. In contrast, excessive Hh pathway activation promotes development of NASH, cirrhosis, and primary liver cancer, the major causes of liver-related deaths. Thus, suppressing excessive Hh pathway activity is a potential approach to prevent progressive liver damage in NAFLD. Various pharmacologic agents that inhibit Hh signaling are available and approved for cancer therapeutics; more are being developed to optimize the benefits and minimize the risks of inhibiting this pathway. In this review we will describe the Hh pathway, summarize the evidence for its role in NAFLD evolution, and discuss the potential role for Hh pathway inhibitors as therapies to prevent NASH, cirrhosis and liver cancer.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- a Division of Gastroenterology, Department of Medicine , Duke University Medical Center , Durham , NC , USA.,b Department of Gastroenterology , Hospital de Santa Maria, CHLN , Lisbon , Portugal
| | - Anna Mae Diehl
- a Division of Gastroenterology, Department of Medicine , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
37
|
Machado MV, Diehl AM. Hedgehog signalling in liver pathophysiology. J Hepatol 2018; 68:550-562. [PMID: 29107151 PMCID: PMC5957514 DOI: 10.1016/j.jhep.2017.10.017] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022]
Abstract
Liver disease remains a leading cause of mortality worldwide despite recent successes in the field of viral hepatitis, because increases in alcohol consumption and obesity are fuelling an epidemic of chronic fatty liver disease for which there are currently no effective medical therapies. About 20% of individuals with chronic liver injury ultimately develop end-stage liver disease due to cirrhosis. Hence, treatments to prevent and reverse cirrhosis in individuals with ongoing liver injury are desperately needed. The development of successful treatments requires an improved understanding of the mechanisms controlling liver disease progression. The liver responds to diverse insults with a conserved wound healing response, suggesting that it might be generally beneficial to optimise pathways that are crucial for effective liver repair. The Hedgehog pathway has emerged as a potential target based on compelling preclinical and clinical data, which demonstrate that it critically regulates the liver's response to injury. Herein, we will summarise evidence of the Hedgehog pathway's role in liver disease and discuss how modulating pathway activity might be applied to improve liver disease outcomes.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA,Gastroenterology Department, Hospital de Santa Maria, CHLN, Lisbon, Portugal
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
38
|
Stagni F, Giacomini A, Emili M, Guidi S, Bartesaghi R. Neurogenesis impairment: An early developmental defect in Down syndrome. Free Radic Biol Med 2018; 114:15-32. [PMID: 28756311 DOI: 10.1016/j.freeradbiomed.2017.07.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023]
Abstract
Down syndrome (DS) is characterized by brain hypotrophy and intellectual disability starting from early life stages. Accumulating evidence shows that the phenotypic features of the DS brain can be traced back to the fetal period since the DS brain exhibits proliferation potency reduction starting from the critical time window of fetal neurogenesis. This defect is worsened by the fact that neural progenitor cells exhibit reduced acquisition of a neuronal phenotype and an increase in the acquisition of an astrocytic phenotype. Consequently, the DS brain has fewer neurons in comparison with the typical brain. Although apoptotic cell death may be increased in DS, this does not seem to be the major cause of brain hypocellularity. Evidence obtained in brains of individuals with DS, DS-derived induced pluripotent stem cells (iPSCs), and DS mouse models has provided some insight into the mechanisms underlying the developmental defects due to the trisomic condition. Although many triplicated genes may be involved, in the light of the studies reviewed here, DYRK1A, APP, RCAN1 and OLIG1/2 appear to be particularly important determinants of many neurodevelopmental alterations that characterize DS because their triplication affects both the proliferation and fate of neural precursor cells as well as apoptotic cell death. Based on the evidence reviewed here, pathways downstream to these genes may represent strategic targets, for the design of possible interventions.
Collapse
Affiliation(s)
- Fiorenza Stagni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
39
|
Bao C, Kramata P, Lee HJ, Suh N. Regulation of Hedgehog Signaling in Cancer by Natural and Dietary Compounds. Mol Nutr Food Res 2017; 62. [PMID: 29164817 DOI: 10.1002/mnfr.201700621] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/14/2017] [Indexed: 12/12/2022]
Abstract
The aberrant Hedgehog (Hh) signaling induced by mutations or overexpression of the signaling mediators has been implicated in cancer, associated with processes including inflammation, tumor cell growth, invasion, and metastasis, as well as cancer stemness. Small molecules targeting the regulatory components of the Hh signaling pathway, especially Smoothened (Smo), have been developed for the treatment of cancer. However, acquired resistance to a Smo inhibitor vismodegib observed in clinical trials suggests that other Hh signaling components need to be explored as potential anticancer targets. Natural and dietary compounds provide a resource for the development of potent agents affecting intracellular signaling cascades, and numerous studies have been conducted to evaluate the efficacy of natural products in targeting the Hh signaling pathway. In this review, we summarize the role of Hh signaling in tumorigenesis, discuss results from recent studies investigating the effect of natural products and dietary components on Hh signaling in cancer, and provide insight on novel small molecules as potential Hh signaling inhibitors.
Collapse
Affiliation(s)
- Cheng Bao
- Department of Food Science and Technology, Chung-Ang University, Anseong, South Korea
| | - Pavel Kramata
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Hong Jin Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong, South Korea
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
40
|
Co-expression analysis revealed PTCH1-3'UTR promoted cell migration and invasion by activating miR-101-3p/SLC39A6 axis in non-small cell lung cancer: implicating the novel function of PTCH1. Oncotarget 2017; 9:4798-4813. [PMID: 29435142 PMCID: PMC5797013 DOI: 10.18632/oncotarget.23219] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/16/2017] [Indexed: 12/25/2022] Open
Abstract
Metastasis is the most common cause of mortality for non-small cell lung cancer (NSCLC). PTCH1, a receptor of Hedgehog (Hh) pathway, is reported to suppress cell proliferation. Interestingly, our previous study showed PTCH1 silencing promoted cell proliferation but inhibited cell migration and invasion of NSCLC cells. However, the precise mechanisms of PTCH1 regulating NSCLC metastasis remain unclear. PTCH1 has multiple splicing variants, which all share the same 3'UTR sequence, meanwhile, emerging studies have shown competing endogenous RNAs (ceRNAs) play important roles in regulating cancer progression. Therefore, we hypothesized the functions of PTCH1-3'UTR in NSCLC in present study to reveal its role as a ceRNA. Here, we find overexpression of PTCH1-3'UTR promotes cell migration, invasion and adhesion, but does not affect cell proliferation in NSCLC cells. By combining weighted correlation network analysis (WGCNA) analysis and experimental validation, we reported PTCH1-3'UTR acted as a sponge to absorb miR-101-3p and promoted SLC39A6 expression. Moreover, we observed low expression of miR-101-3p and PTCH1 and high SLC39A6 levels were positively correlated with NSCLC progression. Therefore, our results help to understand the function of PTCH1 in NSCLC tumorigenesis and provide novel insights for the prevention of NSCLC metastasis.
Collapse
|
41
|
Shen X, Peng Y, Li H. The Injury-Related Activation of Hedgehog Signaling Pathway Modulates the Repair-Associated Inflammation in Liver Fibrosis. Front Immunol 2017; 8:1450. [PMID: 29163520 PMCID: PMC5681491 DOI: 10.3389/fimmu.2017.01450] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022] Open
Abstract
Liver fibrosis is a wound healing response initiated by inflammation responding for different iterative parenchymal damages caused by diverse etiologies. Immune cells, which exert their ability of either inducing injury or promoting repair, have been regarded as crucial participants in the fibrogenic response. A characteristic feature of the fibrotic microenvironment associated with chronic liver injury is aberrant activation of hedgehog (Hh) signaling pathway. Growing evidence from a number of different studies in vivo and in vitro has indicated that immune-mediated events involved in liver fibrogenesis are regulated by Hh signaling pathway. In this review, we emphasize the impacts of injury-activated Hh signaling on liver fibrogenesis through modulating repair-related inflammation and focus on the regulatory action of aberrant Hh signaling on repair-related inflammatory responses mediated by hepatic classical and non-classical immune cell populations in the progression of liver fibrosis. Moreover, we also assess the potentiality of Hh pathway inhibitors as good candidates for anti-fibrotic therapeutic agents because of their immune regulation actions for fibrogenic liver repair. The identification of immune-modulatory mechanisms of Hh signaling pathway underlying the fibrotic process of chronic liver diseases might provide a basis for Hh-centered therapeutic strategies for liver fibrosis.
Collapse
Affiliation(s)
- Xin Shen
- Department of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yu Peng
- Department of Information Engineering, Hubei University of Chinese Medicine, Wuhan, China
| | - Hanmin Li
- Hepatic Disease Institute, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
42
|
Szczepny A, Rogers S, Jayasekara WSN, Park K, McCloy RA, Cochrane CR, Ganju V, Cooper WA, Sage J, Peacock CD, Cain JE, Burgess A, Watkins DN. The role of canonical and non-canonical Hedgehog signaling in tumor progression in a mouse model of small cell lung cancer. Oncogene 2017; 36:5544-5550. [PMID: 28581526 PMCID: PMC5623150 DOI: 10.1038/onc.2017.173] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
Abstract
Hedgehog (Hh) signaling regulates cell fate and self-renewal in development and cancer. Canonical Hh signaling is mediated by Hh ligand binding to the receptor Patched (Ptch), which in turn activates Gli-mediated transcription through Smoothened (Smo), the molecular target of the Hh pathway inhibitors used as cancer therapeutics. Small cell lung cancer (SCLC) is a common, aggressive malignancy with universally poor prognosis. Although preclinical studies have shown that Hh inhibitors block the self-renewal capacity of SCLC cells, the lack of activating pathway mutations have cast doubt over the significance of these observations. In particular, the existence of autocrine, ligand-dependent Hh signaling in SCLC has been disputed. In a conditional Tp53;Rb1 mutant mouse model of SCLC, we now demonstrate a requirement for the Hh ligand Sonic Hedgehog (Shh) for the progression of SCLC. Conversely, we show that conditional Shh overexpression activates canonical Hh signaling in SCLC cells, and markedly accelerates tumor progression. When compared to mouse SCLC tumors expressing an activating, ligand-independent Smo mutant, tumors overexpressing Shh exhibited marked chromosomal instability and Smoothened-independent upregulation of Cyclin B1, a putative non-canonical arm of the Hh pathway. In turn, we show that overexpression of Cyclin B1 induces chromosomal instability in mouse embryonic fibroblasts lacking both Tp53 and Rb1. These results provide strong support for an autocrine, ligand-dependent model of Hh signaling in SCLC pathogenesis, and reveal a novel role for non-canonical Hh signaling through the induction of chromosomal instability.
Collapse
Affiliation(s)
- A Szczepny
- Centre for Cancer Research, The Hudson Institute for Medical Research, Clayton, VIC, Australia
| | - S Rogers
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, UNSW Faculty of Medicine, Sydney, NSW, Australia
| | - W S N Jayasekara
- Centre for Cancer Research, The Hudson Institute for Medical Research, Clayton, VIC, Australia
| | - K Park
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - R A McCloy
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - C R Cochrane
- Centre for Cancer Research, The Hudson Institute for Medical Research, Clayton, VIC, Australia.,School of Clinical Sciences, Faculty of Medicine, Nursing and Health Science, Monash University, Clayton, VIC, Australia
| | - V Ganju
- Centre for Cancer Research, The Hudson Institute for Medical Research, Clayton, VIC, Australia.,School of Clinical Sciences, Faculty of Medicine, Nursing and Health Science, Monash University, Clayton, VIC, Australia.,Department of Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - W A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - J Sage
- Departments of Pediatrics and Genetics, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - C D Peacock
- Department of Translational Hematology Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - J E Cain
- Centre for Cancer Research, The Hudson Institute for Medical Research, Clayton, VIC, Australia
| | - A Burgess
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, UNSW Faculty of Medicine, Sydney, NSW, Australia
| | - D N Watkins
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, UNSW Faculty of Medicine, Sydney, NSW, Australia.,Department of Thoracic Medicine, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
43
|
Ye Y, Wei Y, Xu Y, Li Y, Wang R, Chen J, Zhou Y, Fu Z, Chen Y, Wang X, Yu R, Bai C, Li G, Chen R, Chen T. Induced MiR-1249 expression by aberrant activation of Hedegehog signaling pathway in hepatocellular carcinoma. Exp Cell Res 2017; 355:9-17. [PMID: 28365245 DOI: 10.1016/j.yexcr.2017.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 03/04/2017] [Accepted: 03/07/2017] [Indexed: 12/29/2022]
Abstract
Aberrant activations of Hedegehog (Hh) signaling were found in hepatocellular carcinoma (HCC) and some other cancer types. However, the details have not been completely understood and the underlying mechanism remains unclear. Here we reported that miR-1249 transcription in HCC cells was regulated through direct binding to the conserved sequences in miR-1249 promoter region by Gli1, which functions as a transcription factor and is a component in the Hh signaling pathway. Interestingly, expression of tumor suppressor PTCH1, which is another component of the Hh signaling pathway, was inhibited by miR-1249 through targeting its 3'-untranslated region. Down-regulation of PTCH1 further enhanced the downstream effects mediated by Gli1. In consistent with these findings, miR-1249 expression level was correlated with degree of prognosis (p=0.005) in HCC patients. Taken together, our results suggested the existence of a positive feedback loop comprised of Gli1, miR-1249 and PTCH1. During the process of HCC progression, this positive feedback loop could be continuously activated to enhance tumor cell growth, migration and invasion.
Collapse
Affiliation(s)
- Yibiao Ye
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunping Wei
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunxiuxiu Xu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanshan Li
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruomei Wang
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Zhou
- Department of General Surgery, Guangdong General Hospital, Guangzhou, China
| | - Zhiqiang Fu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Chen
- Pearl Laboratory Animal Sci. & Tech. Co., Ltd., Guangzhou, China
| | - Xin Wang
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA; Hepatoscience Inc., Sunnyvale, CA 94085, USA
| | - Ruiping Yu
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Chunling Bai
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Guangpeng Li
- Key Laboratory of National Education Ministry for Mammalian Reproductive Biology and Biotechnology, Inner Mongolia University, Huhhot, PR China
| | - Rufu Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Tao Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
44
|
Monkkonen T, Landua JD, Visbal AP, Lewis MT. Epithelial and non-epithelial Ptch1 play opposing roles to regulate proliferation and morphogenesis of the mouse mammary gland. Development 2017; 144:1317-1327. [PMID: 28275010 PMCID: PMC5399619 DOI: 10.1242/dev.140434] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Patched 1 (Ptch1) has epithelial, stromal and systemic roles in murine mammary gland organogenesis, yet specific functions remain undefined. Cre-recombinase-mediated Ptch1 ablation in mammary epithelium increased proliferation and branching, but did not phenocopy transgenic expression of activated smoothened (SmoM2). The epithelium showed no evidence of canonical hedgehog signaling, and hyperproliferation was not blocked by smoothened (SMO) inhibition, suggesting a non-canonical function of PTCH1. Consistent with this possibility, nuclear localization of cyclin B1 was increased. In non-epithelial cells, heterozygous Fsp-Cre-mediated Ptch1 ablation increased proliferation and branching, with dysplastic terminal end buds (TEB) and ducts. By contrast, homozygous Ptch1 ablation decreased proliferation and branching, producing stunted ducts filled with luminal cells showing altered ovarian hormone receptor expression. Whole-gland transplantation into wild-type hosts or estrogen/progesterone treatment rescued outgrowth and hormone receptor expression, but not the histological changes. Bone marrow transplantation failed to rescue outgrowth. Ducts of Fsp-Cre;Ptch1fl/fl mice were similar to Fsp-Cre;SmoM2 ducts, but Fsp-Cre;SmoM2 outgrowths were not stunted, suggesting that the histology might be mediated by Smo in the local stroma, with systemic Ptch1 required for ductal outgrowth and proper hormone receptor expression in the mammary epithelium. Summary: Systemic and tissue-specific depletion of patched 1 in epithelial and stromal compartments of the mammary gland defines functions in ductal patterning, proliferation and gene expression.
Collapse
Affiliation(s)
- Teresa Monkkonen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - John D Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Adriana P Visbal
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA .,Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
45
|
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
|
46
|
Kiecker C, Graham A, Logan M. Differential Cellular Responses to Hedgehog Signalling in Vertebrates-What is the Role of Competence? J Dev Biol 2016; 4:E36. [PMID: 29615599 PMCID: PMC5831800 DOI: 10.3390/jdb4040036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 11/24/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
A surprisingly small number of signalling pathways generate a plethora of cellular responses ranging from the acquisition of multiple cell fates to proliferation, differentiation, morphogenesis and cell death. These diverse responses may be due to the dose-dependent activities of signalling factors, or to intrinsic differences in the response of cells to a given signal-a phenomenon called differential cellular competence. In this review, we focus on temporal and spatial differences in competence for Hedgehog (HH) signalling, a signalling pathway that is reiteratively employed in embryos and adult organisms. We discuss the upstream signals and mechanisms that may establish differential competence for HHs in a range of different tissues. We argue that the changing competence for HH signalling provides a four-dimensional framework for the interpretation of the signal that is essential for the emergence of functional anatomy. A number of diseases-including several types of cancer-are caused by malfunctions of the HH pathway. A better understanding of what provides differential competence for this signal may reveal HH-related disease mechanisms and equip us with more specific tools to manipulate HH signalling in the clinic.
Collapse
Affiliation(s)
- Clemens Kiecker
- Department of Developmental Neurobiology, King's College London, Hodgkin Building, Guy's Hospital Campus, London SE1 1UL, UK.
| | - Anthony Graham
- Department of Developmental Neurobiology, King's College London, Hodgkin Building, Guy's Hospital Campus, London SE1 1UL, UK.
| | - Malcolm Logan
- Randall Division of Cell & Molecular Biophysics, King's College London, Hodgkin Building, Guy's Hospital Campus, London SE1 1UL, UK.
| |
Collapse
|
47
|
Cai H, Li H, Li J, Li X, Li Y, Shi Y, Wang D. Sonic hedgehog signaling pathway mediates development of hepatocellular carcinoma. Tumour Biol 2016; 37:16199–16205. [PMID: 27744627 DOI: 10.1007/s13277-016-5463-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/23/2016] [Indexed: 02/04/2023] Open
Abstract
Although abnormal activation of the sonic hedgehog (Shh) signaling pathway has been demonstrated in human hepatocellular carcinoma (HCC) patients and in most HCC cell lines, the mechanism by which the Shh pathway promotes the development of HCC remains uncertain. Using a liver cancer model induced by diethylnitrosamine (DEN) which mimics the process from liver injury, abnormal hepatocyte proliferation, and hepatocirrhosis to hepatocyte canceration, we investigated the abnormal activation of the Shh pathway by examining the expression of Shh, patched-1 (Ptch), smoothened (SMO), and glioma-associated oncogene-1 (Gli1) genes. During this process, the expression of CDK1 and cyclin B1 protein, which are two components of the M-phase promoting factor (MPF) controlling G2/M transition, was also examined to explore the potential relationship between Shh activation and cell cycle progression. We observed that the cells with Shh, Ptch, and Gli1 protein expression were mainly distributed in hyperplastic nodule, cancerous node, the epithelia of interlobular bile duct, and precancerous tissues. A gradually increasing tendency of the positive expression rate of Shh, Ptch, and Gli1 proteins in the process from the beginning normal tissue to the final cancer formation was revealed. The cyclin B1 and CDK1 expression level was higher in the DEN-induced rats as compared with normal rats, and their expression was mainly distributed in the portal area of the liver, hyperplastic nodule, cancerous node, and precancerous tissues. Our results suggested that the Shh signaling pathway is activated during liver carcinogenesis, and activated Shh signaling promotes the cell proliferation by facilitating the G2/M transition through increasing the expression of cyclin B1 and CDK1 protein, which eventually results in the development of liver cancer. Better understanding of the Shh signaling pathway in HCC may contribute to the development of novel therapeutic strategies in inhibiting cell proliferation and promoting cell cycle arrest.
Collapse
Affiliation(s)
- Heng Cai
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Hongxing Li
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Jingmin Li
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Xiaoyan Li
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Yana Li
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Yan Shi
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China
| | - Dong Wang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, 264003, People's Republic of China.
| |
Collapse
|
48
|
Fleet A, Lee JPY, Tamachi A, Javeed I, Hamel PA. Activities of the Cytoplasmic Domains of Patched-1 Modulate but Are Not Essential for the Regulation of Canonical Hedgehog Signaling. J Biol Chem 2016; 291:17557-68. [PMID: 27325696 DOI: 10.1074/jbc.m116.731745] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
The Hedgehog (Hh) pathway is a highly conserved signaling cascade crucial for cell fate determination during embryogenesis. Response to the Hh ligands is mediated by the receptor Patched-1 (Ptch1), a 12-pass transmembrane glycoprotein. Despite its essential role in Hh signaling and its activity as a tumor suppressor, Ptch1 remains largely uncharacterized. We demonstrate here that Ptch1 binds to itself to form oligomeric structures. Oligomerization is mediated by two distinct, structurally disordered, intracellular domains spanning amino acids 584-734 ("middle loop") and 1162-1432 (C terminus). However, oligomerization is not required for Ptch1-dependent regulation of the canonical Hh pathway operating through Smo. Expression of a mutant protein that deletes both regions represses the Hh pathway and responds to the addition of Hh ligand independent of its inability to bind other factors such as Smurf2. Additionally, deletion of the cytoplasmic middle loop domain generates a Ptch1 mutant that, despite binding to Hh ligand, constitutively suppresses Hh signaling and increases the length of primary cilia. Constitutive activity because of deletion of this region is reversed by further deletion of specific sequences in the cytoplasmic C-terminal domain. These data reveal an interaction between the cytoplasmic domains of Ptch1 and that these domains modulate Ptch1 activity but are not essential for regulation of the Hh pathway.
Collapse
Affiliation(s)
- Andrew Fleet
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jennifer P Y Lee
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Aaliya Tamachi
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Imaan Javeed
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Paul A Hamel
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
49
|
Verdelho Machado M, Diehl AM. Role of Hedgehog Signaling Pathway in NASH. Int J Mol Sci 2016; 17:E857. [PMID: 27258259 PMCID: PMC4926391 DOI: 10.3390/ijms17060857] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/18/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the number one cause of chronic liver disease in the Western world. Although only a minority of patients will ultimately develop end-stage liver disease, it is not yet possible to efficiently predict who will progress and, most importantly, effective treatments are still unavailable. Better understanding of the pathophysiology of this disease is necessary to improve the clinical management of NAFLD patients. Epidemiological data indicate that NAFLD prognosis is determined by an individual's response to lipotoxic injury, rather than either the severity of exposure to lipotoxins, or the intensity of liver injury. The liver responds to injury with a synchronized wound-healing response. When this response is abnormal, it leads to pathological scarring, resulting in progressive fibrosis and cirrhosis, rather than repair. The hedgehog pathway is a crucial player in the wound-healing response. In this review, we summarize the pre-clinical and clinical evidence, which demonstrate the role of hedgehog pathway dysregulation in NAFLD pathogenesis, and the preliminary data that place the hedgehog pathway as a potential target for the treatment of this disease.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
- Gastroenterology Department, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte (CHLN), Lisboa 1649-035, Portugal.
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
50
|
Utheim TP, Islam R, Fostad IG, Eidet JR, Sehic A, Olstad OK, Dartt DA, Messelt EB, Griffith M, Pasovic L. Storage Temperature Alters the Expression of Differentiation-Related Genes in Cultured Oral Keratinocytes. PLoS One 2016; 11:e0152526. [PMID: 27023475 PMCID: PMC4811429 DOI: 10.1371/journal.pone.0152526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/15/2016] [Indexed: 12/17/2022] Open
Abstract
Purpose Storage of cultured human oral keratinocytes (HOK) allows for transportation of cultured transplants to eye clinics worldwide. In a previous study, one-week storage of cultured HOK was found to be superior with regard to viability and morphology at 12°C compared to 4°C and 37°C. To understand more of how storage temperature affects cell phenotype, gene expression of HOK before and after storage at 4°C, 12°C, and 37°C was assessed. Materials and Methods Cultured HOK were stored in HEPES- and sodium bicarbonate-buffered Minimum Essential Medium at 4°C, 12°C, and 37°C for one week. Total RNA was isolated and the gene expression profile was determined using DNA microarrays and analyzed with Partek Genomics Suite software and Ingenuity Pathway Analysis. Differentially expressed genes (fold change > 1.5 and P < 0.05) were identified by one-way ANOVA. Key genes were validated using qPCR. Results Gene expression of cultures stored at 4°C and 12°C clustered close to the unstored control cultures. Cultures stored at 37°C displayed substantial change in gene expression compared to the other groups. In comparison with 12°C, 2,981 genes were differentially expressed at 37°C. In contrast, only 67 genes were differentially expressed between the unstored control and the cells stored at 12°C. The 12°C and 37°C culture groups differed most significantly with regard to the expression of differentiation markers. The Hedgehog signaling pathway was significantly downregulated at 37°C compared to 12°C. Conclusion HOK cultures stored at 37°C showed considerably larger changes in gene expression compared to unstored cells than cultured HOK stored at 4°C and 12°C. The changes observed at 37°C consisted of differentiation of the cells towards a squamous epithelium-specific phenotype. Storing cultured ocular surface transplants at 37°C is therefore not recommended. This is particularly interesting as 37°C is the standard incubation temperature used for cell culture.
Collapse
Affiliation(s)
- Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Vestre Viken HF Trust, Drammen, Norway
- Faculty of Health Sciences, National Centre for Optics, Vision and Eye Care, Buskerud and Vestfold University College, Kongsberg, Norway
| | - Rakibul Islam
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Ida G. Fostad
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Jon R. Eidet
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | - Amer Sehic
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Ole K. Olstad
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Darlene A. Dartt
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Edward B. Messelt
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - May Griffith
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lara Pasovic
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|