1
|
Lamson DR, Tarpley M, Addo K, Ji X, Abu Rabe D, Ehe B, Hughes M, Smith GR, Daye LR, Musso DL, Zheng W, Williams KP. Identification of small molecule antagonists of sonic hedgehog/heparin binding with activity in hedgehog functional assays. Biochim Biophys Acta Gen Subj 2024; 1868:130692. [PMID: 39151833 PMCID: PMC11486593 DOI: 10.1016/j.bbagen.2024.130692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Sonic hedgehog (Shh) is a morphogen with important roles in embryonic development and in the development of a number of cancers. Its activity is modulated by interactions with binding partners and co-receptors including heparin and heparin sulfate proteoglycans (HSPG). To identify antagonists of Shh/heparin binding, a diverse collection of 34,560 chemicals was screened in single point 384-well format. We identified and confirmed twenty six novel small molecule antagonists with diverse structures including four scaffolds that gave rise to multiple hits. Nineteen of the confirmed hits blocked binding of the N-terminal fragment of Shh (ShhN) to heparin with IC50 values < 50 μM. In the Shh-responsive C3H10T1/2 cell model, four of the compounds demonstrated the ability to block ShhN-induced alkaline phosphatase activity. To demonstrate a direct and selective effect on ShhN ligand mediated activity, two of the compounds were able to block induction of Gli1 mRNA, a primary downstream marker for Shh signaling activity, in Shh-mediated but not Smoothened agonist (SAG)-mediated C3H10T1/2 cells. Direct binding of the two compounds to ShhN was confirmed by thermal shift assay and molecular docking simulations, with both compounds docking with the N-terminal heparin binding domain of Shh. Overall, our findings indicate that small molecule compounds that block ShhN binding to heparin and act to inhibit Shh mediated activity in vitro can be identified. We propose that the interaction between Shh and HSPGs provides a novel target for identifying small molecules that bind Shh, potentially leading to novel tool compounds to probe Shh ligand function.
Collapse
Affiliation(s)
- David R Lamson
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Michael Tarpley
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Kezia Addo
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Xiaojia Ji
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Dina Abu Rabe
- Biomanufacturing Research Institute and Technology Enterprise, USA; INBS PhD Program, USA
| | - Ben Ehe
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Mark Hughes
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Ginger R Smith
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Laura R Daye
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - David L Musso
- Biomanufacturing Research Institute and Technology Enterprise, USA
| | - Weifan Zheng
- Biomanufacturing Research Institute and Technology Enterprise, USA; Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Kevin P Williams
- Biomanufacturing Research Institute and Technology Enterprise, USA; Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
2
|
Tshilate TS, Ishengoma E, Rhode C. Construction of a high-density linkage map and QTL detection for growth traits in South African abalone (Haliotis midae). Anim Genet 2024; 55:744-760. [PMID: 38945682 DOI: 10.1111/age.13462] [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: 10/02/2023] [Revised: 03/23/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
Haliotis midae is one of the most important molluscs in South African commercial aquaculture. In this study, a high-resolution integrated linkage map was constructed, and QTL identified using 2b-RADseq for genotyping SNPs in three families. The final integrated linkage map was composed by merging the individual family maps, resulting in 3290 informative SNPs mapping to 18 linkage groups, conforming to the known haploid chromosome number for H. midae. The total map spanned 1798.25 cM with an average marker interval of 0.55 cM, representing a genome coverage of 98.76%. QTL analysis, across all three families, resulted in a total of five QTL identified for growth-related traits, shell width, shell length, and total body weight. For shell width and total body weight, one QTL was identified for each trait respectively, whilst three QTL were identified for shell length. The identified QTL respectively explained between 7.20% and 11.40% of the observed phenotypic variance. All three traits were significantly correlated (r = 0.862-0.970; p < 0.01) and shared overlapping QTL. The QTL for growth traits were mapped back to the H. midae draft genome and BLAST searches revealed the identity of candidate genes, such as egf-1, megf10, megf6, tnx, sevp1, kcp, notch1, and scube2 with possible functional roles in H. midae growth. The constructed high-density linkage map and mapped QTL have given valuable insights regarding the genetic architecture of growth-related traits and will be important genetic resources for marker-assisted selection. It remains, however, important to validate causal variants through linkage disequilibrium fine mapping in future.
Collapse
Affiliation(s)
| | - Edson Ishengoma
- Department of Genetics, Stellenbosch University, Matieland, South Africa
- Mkwawa University College of Education, University of Dar es Salaam, Iringa, Tanzania
| | - Clint Rhode
- Department of Genetics, Stellenbosch University, Matieland, South Africa
| |
Collapse
|
3
|
Ehring K, Ehlers SF, Froese J, Gude F, Puschmann J, Grobe K. Two-way Dispatched function in Sonic hedgehog shedding and transfer to high-density lipoproteins. eLife 2024; 12:RP86920. [PMID: 39297609 PMCID: PMC11412720 DOI: 10.7554/elife.86920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
The Sonic hedgehog (Shh) signaling pathway controls embryonic development and tissue homeostasis after birth. This requires regulated solubilization of dual-lipidated, firmly plasma membrane-associated Shh precursors from producing cells. Although it is firmly established that the resistance-nodulation-division transporter Dispatched (Disp) drives this process, it is less clear how lipidated Shh solubilization from the plasma membrane is achieved. We have previously shown that Disp promotes proteolytic solubilization of Shh from its lipidated terminal peptide anchors. This process, termed shedding, converts tightly membrane-associated hydrophobic Shh precursors into delipidated soluble proteins. We show here that Disp-mediated Shh shedding is modulated by a serum factor that we identify as high-density lipoprotein (HDL). In addition to serving as a soluble sink for free membrane cholesterol, HDLs also accept the cholesterol-modified Shh peptide from Disp. The cholesteroylated Shh peptide is necessary and sufficient for Disp-mediated transfer because artificially cholesteroylated mCherry associates with HDL in a Disp-dependent manner, whereas an N-palmitoylated Shh variant lacking C-cholesterol does not. Disp-mediated Shh transfer to HDL is completed by proteolytic processing of the palmitoylated N-terminal membrane anchor. In contrast to dual-processed soluble Shh with moderate bioactivity, HDL-associated N-processed Shh is highly bioactive. We propose that the purpose of generating different soluble forms of Shh from the dual-lipidated precursor is to tune cellular responses in a tissue-type and time-specific manner.
Collapse
Affiliation(s)
- Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | | | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Janna Puschmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of MünsterMünsterGermany
| |
Collapse
|
4
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
5
|
Ehlers SF, Manikowski D, Steffes G, Ehring K, Gude F, Grobe K. A Residual N-Terminal Peptide Enhances Signaling of Depalmitoylated Hedgehog to the Patched Receptor. J Dev Biol 2024; 12:11. [PMID: 38651456 PMCID: PMC11036296 DOI: 10.3390/jdb12020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
During their biosynthesis, Sonic hedgehog (Shh) morphogens are covalently modified by cholesterol at the C-terminus and palmitate at the N-terminus. Although both lipids initially anchor Shh to the plasma membrane of producing cells, it later translocates to the extracellular compartment to direct developmental fates in cells expressing the Patched (Ptch) receptor. Possible release mechanisms for dually lipidated Hh/Shh into the extracellular compartment are currently under intense debate. In this paper, we describe the serum-dependent conversion of the dually lipidated cellular precursor into a soluble cholesteroylated variant (ShhC) during its release. Although ShhC is formed in a Dispatched- and Scube2-dependent manner, suggesting the physiological relevance of the protein, the depalmitoylation of ShhC during release is inconsistent with the previously postulated function of N-palmitate in Ptch receptor binding and signaling. Therefore, we analyzed the potency of ShhC to induce Ptch-controlled target cell transcription and differentiation in Hh-sensitive reporter cells and in the Drosophila eye. In both experimental systems, we found that ShhC was highly bioactive despite the absence of the N-palmitate. We also found that the artificial removal of N-terminal peptides longer than eight amino acids inactivated the depalmitoylated soluble proteins in vitro and in the developing Drosophila eye. These results demonstrate that N-depalmitoylated ShhC requires an N-peptide of a defined minimum length for its signaling function to Ptch.
Collapse
Affiliation(s)
- Sophia F. Ehlers
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Georg Steffes
- Institute for Neuro- and Behavioral Biology, Faculty of Biology, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany;
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| |
Collapse
|
6
|
Zhang Y, Beachy PA. Cellular and molecular mechanisms of Hedgehog signalling. Nat Rev Mol Cell Biol 2023; 24:668-687. [PMID: 36932157 DOI: 10.1038/s41580-023-00591-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 03/19/2023]
Abstract
The Hedgehog signalling pathway has crucial roles in embryonic tissue patterning, postembryonic tissue regeneration, and cancer, yet aspects of Hedgehog signal transmission and reception have until recently remained unclear. Biochemical and structural studies surprisingly reveal a central role for lipids in Hedgehog signalling. The signal - Hedgehog protein - is modified by cholesterol and palmitate during its biogenesis, thereby necessitating specialized proteins such as the transporter Dispatched and several lipid-binding carriers for cellular export and receptor engagement. Additional lipid transactions mediate response to the Hedgehog signal, including sterol activation of the transducer Smoothened. Access of sterols to Smoothened is regulated by the apparent sterol transporter and Hedgehog receptor Patched, whose activity is blocked by Hedgehog binding. Alongside these lipid-centric mechanisms and their relevance to pharmacological pathway modulation, we discuss emerging roles of Hedgehog pathway activity in stem cells or their cellular niches, with translational implications for regeneration and restoration of injured or diseased tissues.
Collapse
Affiliation(s)
- Yunxiao Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute and Neuroscience Department, The Scripps Research Institute, La Jolla, CA, USA
| | - Philip A Beachy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Wu Q, Tian P, He D, Jia Z, He Y, Luo W, Lv X, Wang Y, Zhang P, Liang Y, Zhao W, Qin J, Su P, Jiang YZ, Shao ZM, Yang Q, Hu G. SCUBE2 mediates bone metastasis of luminal breast cancer by modulating immune-suppressive osteoblastic niches. Cell Res 2023; 33:464-478. [PMID: 37142671 PMCID: PMC10235122 DOI: 10.1038/s41422-023-00810-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/02/2023] [Indexed: 05/06/2023] Open
Abstract
Estrogen receptor (ER)-positive luminal breast cancer is a subtype with generally lower risk of metastasis to most distant organs. However, bone recurrence occurs preferentially in luminal breast cancer. The mechanisms of this subtype-specific organotropism remain elusive. Here we show that an ER-regulated secretory protein SCUBE2 contributes to bone tropism of luminal breast cancer. Single-cell RNA sequencing analysis reveals osteoblastic enrichment by SCUBE2 in early bone-metastatic niches. SCUBE2 facilitates release of tumor membrane-anchored SHH to activate Hedgehog signaling in mesenchymal stem cells, thus promoting osteoblast differentiation. Osteoblasts deposit collagens to suppress NK cells via the inhibitory LAIR1 signaling and promote tumor colonization. SCUBE2 expression and secretion are associated with osteoblast differentiation and bone metastasis in human tumors. Targeting Hedgehog signaling with Sonidegib and targeting SCUBE2 with a neutralizing antibody both effectively suppress bone metastasis in multiple metastasis models. Overall, our findings provide a mechanistic explanation for bone preference in luminal breast cancer metastasis and new approaches for metastasis treatment.
Collapse
Affiliation(s)
- Qiuyao Wu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pu Tian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dasa He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenchang Jia
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yunfei He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenqian Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xianzhe Lv
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajun Liang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjin Zhao
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | - Jun Qin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Ji'nan, Shandong, China.
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
Lin YC, Sahoo BK, Gau SS, Yang RB. The biology of SCUBE. J Biomed Sci 2023; 30:33. [PMID: 37237303 PMCID: PMC10214685 DOI: 10.1186/s12929-023-00925-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The SCUBE [Signal peptide-Complement C1r/C1s, Uegf, Bmp1 (CUB)-Epithelial growth factor domain-containing protein] family consists of three proteins in vertebrates, SCUBE1, 2 and 3, which are highly conserved in zebrafish, mice and humans. Each SCUBE gene encodes a polypeptide of approximately 1000 amino acids that is organized into five modular domains: (1) an N-terminal signal peptide sequence, (2) nine tandem epidermal growth factor (EGF)-like repeats, (3) a large spacer region, (4) three cysteine-rich (CR) motifs, and (5) a CUB domain at the C-terminus. Murine Scube genes are expressed individually or in combination during the development of various tissues, including those in the central nervous system and the axial skeleton. The cDNAs of human SCUBE orthologs were originally cloned from vascular endothelial cells, but SCUBE expression has also been found in platelets, mammary ductal epithelium and osteoblasts. Both soluble and membrane-associated SCUBEs have been shown to play important roles in physiology and pathology. For instance, upregulation of SCUBEs has been reported in acute myeloid leukemia, breast cancer and lung cancer. In addition, soluble SCUBE1 is released from activated platelets and can be used as a clinical biomarker for acute coronary syndrome and ischemic stroke. Soluble SCUBE2 enhances distal signaling by facilitating the secretion of dual-lipidated hedgehog from nearby ligand-producing cells in a paracrine manner. Interestingly, the spacer regions and CR motifs can increase or enable SCUBE binding to cell surfaces via electrostatic or glycan-lectin interactions. As such, membrane-associated SCUBEs can function as coreceptors that enhance the signaling activity of various serine/threonine kinase or tyrosine kinase receptors. For example, membrane-associated SCUBE3 functions as a coreceptor that promotes signaling in bone morphogenesis. In humans, SCUBE3 mutations are linked to abnormalities in growth and differentiation of both bones and teeth. In addition to studies on human SCUBE function, experimental results from genetically modified mouse models have yielded important insights in the field of systems biology. In this review, we highlight novel molecular discoveries and critical directions for future research on SCUBE proteins in the context of cancer, skeletal disease and cardiovascular disease.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Binay K Sahoo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shiang-Shin Gau
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan.
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Zheng G, Ren J, Shang L, Bao Y. Sonic Hedgehog Signaling Pathway: A Role in Pain Processing. Neurochem Res 2023; 48:1611-1630. [PMID: 36738366 DOI: 10.1007/s11064-023-03864-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023]
Abstract
Pain, as one of the most prevalent clinical symptoms, is a complex physiological and psychological activity. Long-term severe pain can become unbearable to the body. However, existing treatments do not provide satisfactory results. Therefore, new mechanisms and therapeutic targets need to be urgently explored for pain management. The Sonic hedgehog (Shh) signaling pathway is crucial in embryonic development, cell differentiation and proliferation, and nervous system regulation. Here, we review the recent studies on the Shh signaling pathway and its action in multiple pain-related diseases. The Shh signaling pathway is dysregulated under various pain conditions, such as pancreatic cancer pain, bone cancer pain, chronic post-thoracotomy pain, pain caused by degenerative lumbar disc disease, and toothache. Further studies on the Shh signaling pathway may provide new therapeutic options for pain patients.
Collapse
Affiliation(s)
- Guangda Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing, 100053, China
| | - Juanxia Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning Province, China
| | - Lu Shang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning Province, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
10
|
Kumar S, Prajapati KS, Gupta S. The Multifaceted Role of Signal Peptide-CUB-EGF Domain-Containing Protein (SCUBE) in Cancer. Int J Mol Sci 2022; 23:ijms231810577. [PMID: 36142489 PMCID: PMC9503623 DOI: 10.3390/ijms231810577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 02/05/2023] Open
Abstract
Signal peptide, CUB, and EGF-like domain-containing proteins (SCUBE) are secretory cell surface glycoproteins that play key roles in the developmental process. SCUBE proteins participate in the progression of several diseases, including cancer, and are recognized for their oncogenic and tumor suppressor functions depending on the cellular context. SCUBE proteins promote cancer cell proliferation, angiogenesis, invasion, or metastasis, stemness or self-renewal, and drug resistance. The association of SCUBE with other proteins alters the expression of signaling pathways, including Hedgehog, Notch, TGF-β/Smad2/3, and β-catenin. Further, SCUBE proteins function as potential prognostic and diagnostic biomarkers for breast cancer, renal cell carcinoma, endometrial carcinoma, and nasopharyngeal carcinoma. This review presents key features of SCUBE family members, and their structure and functions, and highlights their contribution in the development and progression of cancer. A comprehensive understanding of the role of SCUBE family members offers novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda 151401, India
| | - Kumari Sunita Prajapati
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda 151401, India
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, OH 44106, USA
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, OH 44106, USA
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-368-6162; Fax: +1-216-368-0213
| |
Collapse
|
11
|
He Y, Wang J, Jiang X, Gao J, Cheng Y, Liang T, Zhou J, Sun L, Zhang G. Effects of an inhibitor of the SHH signaling pathway on endometrial cells of patients with endometriosis. BMC Mol Cell Biol 2022; 23:37. [PMID: 35933378 PMCID: PMC9356504 DOI: 10.1186/s12860-022-00426-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Endometriosis is one of the most common gynecological diseases, and seriously reduces the quality of life of patients. However, the pathogenesis of this disease is unclear. Therefore, more studies are needed to elucidate its pathogenesis. Our previous publication found that the Sonic Hedgehog (SHH) signaling pathway was activated in endometriosis. This study tested whether SHH signaling in endometrial stromal cells (ESCs) was critical for the pathogenesis of endometriosis. METHODS To examine the effect of inhibiting the SHH signaling pathway on endometriosis, we first isolated ESCs from eutopic endometrial tissues of patients with or without endometriosis and identified the extracted cells by morphological observation and immunofluorescence. Then, we treated ESCs with the GLI inhibitor GANT61 and used CCK-8, wound healing and invasion assays to detect cell activities, such as proliferation, invasion and metastasis. Furthermore, we detected the expression of key proteins and proliferation markers of the SHH signaling pathway in the lesions of nude mice using immunochemistry. RESULTS We demonstrated that higher concentrations of GANT61 decreased the proliferation rate and migration distance of ESCs. We observed that GANT61 inhibited the invasion of ESCs. In addition, blockage of the SHH signaling pathway significantly reduced cell proliferation in vitro. CONCLUSIONS Our study suggested that inhibition of the SHH pathway is involved in cell proliferation and invasive growth in the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Yanan He
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - J Wang
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinyan Jiang
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianhua Gao
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Cheng
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tian Liang
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jun Zhou
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liyuan Sun
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guangmei Zhang
- Department of Gynaecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
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: 18] [Impact Index Per Article: 6.0] [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
|
Dispatching plasma membrane cholesterol and Sonic Hedgehog dispatch: two sides of the same coin? Biochem Soc Trans 2021; 49:2455-2463. [PMID: 34515747 PMCID: PMC8589413 DOI: 10.1042/bst20210918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Vertebrate and invertebrate Hedgehog (Hh) morphogens signal over short and long distances to direct cell fate decisions during development and to maintain tissue homeostasis after birth. One of the most important questions in Hh biology is how such Hh signaling to distant target cells is achieved, because all Hh proteins are secreted as dually lipidated proteins that firmly tether to the outer plasma membrane leaflet of their producing cells. There, Hhs multimerize into light microscopically visible storage platforms that recruit factors required for their regulated release. One such recruited release factor is the soluble glycoprotein Scube2 (Signal sequence, cubulin domain, epidermal-growth-factor-like protein 2), and maximal Scube2 function requires concomitant activity of the resistance-nodulation-division (RND) transporter Dispatched (Disp) at the plasma membrane of Hh-producing cells. Although recently published cryo-electron microscopy-derived structures suggest possible direct modes of Scube2/Disp-regulated Hh release, the mechanism of Disp-mediated Hh deployment is still not fully understood. In this review, we discuss suggested direct modes of Disp-dependent Hh deployment and relate them to the structural similarities between Disp and the related RND transporters Patched (Ptc) and Niemann-Pick type C protein 1. We then discuss open questions and perspectives that derive from these structural similarities, with particular focus on new findings that suggest shared small molecule transporter functions of Disp to deplete the plasma membrane of cholesterol and to modulate Hh release in an indirect manner.
Collapse
|
14
|
Manikowski D, Ehring K, Gude F, Jakobs P, Froese J, Grobe K. Hedgehog lipids: Promotors of alternative morphogen release and signaling?: Conflicting findings on lipidated Hedgehog transport and signaling can be explained by alternative regulated mechanisms to release the morphogen. Bioessays 2021; 43:e2100133. [PMID: 34611914 DOI: 10.1002/bies.202100133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022]
Abstract
Two posttranslational lipid modifications present on all Hedgehog (Hh) morphogens-an N-terminal palmitate and a C-terminal cholesterol-are established and essential regulators of Hh biofunction. Yet, for several decades, the question of exactly how both lipids contribute to Hh signaling remained obscure. Recently, cryogenic electron microscopy revealed different modes by which one or both lipids may contribute directly to Hh binding and signaling to its receptor Patched1 (Ptc). Some of these modes demand that the established release factor Dispatched1 (Disp) extracts dual-lipidated Hh from the cell surface, and that another known upstream signaling modulator called Scube2 chaperones the dual-lipidated morphogen to Ptc. By mechanistically and biochemically aligning this concept with established in vivo and recent in vitro findings, this reflection identifies remaining questions in lipidated Hh transport and evaluates additional mechanisms of Disp- and Scube2-regulated release of a second bioactive Hh fraction that has one or both lipids removed.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, North Rhine-Westphalia, Germany
| |
Collapse
|
15
|
Ehring K, Manikowski D, Goretzko J, Froese J, Gude F, Jakobs P, Rescher U, Kirchhefer U, Grobe K. Conserved cholesterol-related activities of Dispatched 1 drive Sonic hedgehog shedding from the cell membrane. J Cell Sci 2021; 135:271842. [PMID: 34308968 PMCID: PMC8403983 DOI: 10.1242/jcs.258672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/08/2021] [Indexed: 01/04/2023] Open
Abstract
The Sonic hedgehog (Shh) pathway controls embryonic development and tissue homeostasis after birth. Long-standing questions about this pathway include how the dual-lipidated, firmly plasma membrane-associated Shh ligand is released from producing cells to signal to distant target cells and how the resistance-nodulation-division transporter Dispatched 1 (Disp, also known as Disp1) regulates this process. Here, we show that inactivation of Disp in Shh-expressing human cells impairs proteolytic Shh release from its lipidated terminal peptides, a process called ectodomain shedding. We also show that cholesterol export from Disp-deficient cells is reduced, that these cells contain increased cholesterol amounts in the plasma membrane, and that Shh shedding from Disp-deficient cells is restored by pharmacological membrane cholesterol extraction and by overexpression of transgenic Disp or the structurally related protein Patched 1 (Ptc, also known as Ptch1; a putative cholesterol transporter). These data suggest that Disp can regulate Shh function via controlled cell surface shedding and that membrane cholesterol-related molecular mechanisms shared by Disp and Ptc exercise such sheddase control.
Collapse
Affiliation(s)
- Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Jonas Goretzko
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Ursula Rescher
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of Münster, Domagkstrasse 12, D-48149 Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| |
Collapse
|
16
|
Association of Sonic Hedgehog with the extracellular matrix requires its zinc-coordination center. BMC Mol Cell Biol 2021; 22:22. [PMID: 33863273 PMCID: PMC8052667 DOI: 10.1186/s12860-021-00359-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Sonic Hedgehog (Shh) has a catalytic cleft characteristic for zinc metallopeptidases and has significant sequence similarities with some bacterial peptidoglycan metallopeptidases defining a subgroup within the M15A family that, besides having the characteristic zinc coordination motif, can bind two calcium ions. Extracellular matrix (ECM) components in animals include heparan-sulfate proteoglycans, which are analogs of bacterial peptidoglycan and are involved in the extracellular distribution of Shh. Results We found that the zinc-coordination center of Shh is required for its association to the ECM as well as for non-cell autonomous signaling. Association with the ECM requires the presence of at least 0.1 μM zinc and is prevented by mutations affecting critical conserved catalytical residues. Consistent with the presence of a conserved calcium binding domain, we find that extracellular calcium inhibits ECM association of Shh. Conclusions Our results indicate that the putative intrinsic peptidase activity of Shh is required for non-cell autonomous signaling, possibly by enzymatically altering ECM characteristics. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00359-5.
Collapse
|
17
|
Tsao KC, Lin YC, Chen YT, Lai SL, Yang RB. Zebrafish scube1 and scube2 cooperate in promoting Vegfa signaling during embryonic vascularization. Cardiovasc Res 2021; 118:1074-1087. [PMID: 33788916 DOI: 10.1093/cvr/cvab125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 11/18/2020] [Accepted: 03/30/2021] [Indexed: 11/14/2022] Open
Abstract
AIMS The secreted and membrane-anchored SCUBE (signal peptide-CUB-EGF domain-containing proteins) gene family composed of 3 members was originally identified from endothelial cells (ECs). We recently showed that membrane SCUBE2 binds vascular endothelial growth factor A (VEGFA) and acts as a co-receptor for VEGF receptor 2 (VEGFR2) to modulate EC migration, proliferation and tube formation during postnatal and tumor angiogenesis. However, whether these SCUBE genes cooperate in modulating VEGF signaling during embryonic vascular development remains unknown. METHODS AND RESULTS To further dissect the genetic interactions of these scube genes, transcription activator-like effector nuclease-mediated genome editing was used to generate knockout (KO) alleles of each scube gene. No overt vascular phenotypes were seen in any single scube KO mutants because of compensation by other scube genes during zebrafish development. However, scube1 and scube2 double KO (DKO) severely impaired EC filopodia extensions, migration, and proliferation, thus disrupting proper vascular lumen formation during vasculogenesis and angiogenesis as well as development of the organ-specific intestinal vasculature. Further genetic, biochemical, and molecular analyses revealed that Scube1 and Scube2 might act cooperatively at the cell-surface receptor level to facilitate Vegfa signaling during zebrafish embryonic vascularization. CONCLUSIONS We showed for the first time that cooperation between scube1 and scube2 is critical for proper regulation of angiogenic cell behaviors and formation of functional vessels during zebrafish embryonic development. TRANSLATIONAL PERSPECTIVE Our studies indicate that targeting SCUBE1 and/or SCUBE2 on modulating VEGF signaling might provide potential therapeutic treatments or VEGF-mediated proliferative pathological vascular diseases.
Collapse
Affiliation(s)
- Ku-Chi Tsao
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuh-Charn Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ting Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
18
|
Anusha, Dalal H, Subramanian S, V P S, Gowda DA, H K, Damodar S, Vyas N. Exovesicular-Shh confers Imatinib resistance by upregulating Bcl2 expression in chronic myeloid leukemia with variant chromosomes. Cell Death Dis 2021; 12:259. [PMID: 33707419 PMCID: PMC7952724 DOI: 10.1038/s41419-021-03542-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 01/29/2023]
Abstract
Chronic myeloid leukemia (CML) patients with complex chromosomal translocations as well as non-compliant CML patients often demonstrate short-lived responses and poor outcomes on the current therapeutic regimes using Imatinib and its variants. It has been derived so far that leukemic stem cells (LSCs) are responsible for Imatinib resistance and CML progression. Sonic hedgehog (Shh) signaling has been implicated in proliferation of this Imatinib-resistant CD34(+) LSCs. Our work here identifies the molecular mechanism of Shh-mediated mutation-independent Imatinib resistance that is most relevant for treating CML-variants and non-compliant patients. Our results elucidate that while Shh can impart stemness, it also upregulates expression of anti-apoptotic protein—Bcl2. It is the upregulation of Bcl2 that is involved in conferring Imatinib resistance to the CD34(+) LSCs. Sub-toxic doses of Bcl2 inhibitor or Shh inhibitor (<<IC50), when used as adjuvants along with Imatinib, can re-sensitize Shh signaling cells to Imatinib. Our work here highlights the need to molecularly stratify CML patients and implement combinatorial therapy to overcome the current limitations and improve outcomes in CML.
Collapse
Affiliation(s)
- Anusha
- Manipal Academy of Higher Education (MAHE), Manipal, 576104, India.,St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, 560034, India
| | - Hamza Dalal
- Mazumdar Shaw Medical Center, Narayana Health City, Bangalore, 560099, India
| | - Sitalakshmi Subramanian
- St. John's Medical College and Hosptial, St. John's Academy of Health Sciences, Bangalore, 560034, India
| | - Snijesh V P
- St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, 560034, India
| | - Divya A Gowda
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS-GKVK Campus, Bellary Road, Bangalore, 560065, India
| | - Krishnamurthy H
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, UAS-GKVK Campus, Bellary Road, Bangalore, 560065, India
| | - Sharat Damodar
- Mazumdar Shaw Medical Center, Narayana Health City, Bangalore, 560099, India.
| | - Neha Vyas
- St. John's Research Institute, St. John's Academy of Health Sciences, Bangalore, 560034, India.
| |
Collapse
|
19
|
Manikowski D, Kastl P, Schürmann S, Ehring K, Steffes G, Jakobs P, Grobe K. C-Terminal Peptide Modifications Reveal Direct and Indirect Roles of Hedgehog Morphogen Cholesteroylation. Front Cell Dev Biol 2021; 8:615698. [PMID: 33511123 PMCID: PMC7835520 DOI: 10.3389/fcell.2020.615698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/03/2020] [Indexed: 01/20/2023] Open
Abstract
Hedgehog (Hh) morphogens are involved in embryonic development and stem cell biology and, if misregulated, can contribute to cancer. One important post-translational modification with profound impact on Hh biofunction is its C-terminal cholesteroylation during biosynthesis. The current hypothesis is that the cholesterol moiety is a decisive factor in Hh association with the outer plasma membrane leaflet of producing cells, cell-surface Hh multimerization, and its transport and signaling. Yet, it is not decided whether the cholesterol moiety is directly involved in all of these processes, because their functional interdependency raises the alternative possibility that the cholesterol initiates early processes directly and that these processes can then steer later stages of Hh signaling independent of the lipid. We generated variants of the C-terminal Hh peptide and observed that these cholesteroylated peptides variably impaired several post-translational processes in producing cells and Hh biofunction in Drosophila melanogaster eye and wing development. We also found that substantial Hh amounts separated from cholesteroylated peptide tags in vitro and in vivo and that tagged and untagged Hh variants lacking their C-cholesterol moieties remained bioactive. Our approach thus confirms that Hh cholesteroylation is essential during the early steps of Hh production and maturation but also suggests that it is dispensable for Hh signal reception at receiving cells.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Georg Steffes
- Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and the Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
20
|
Wierbowski BM, Petrov K, Aravena L, Gu G, Xu Y, Salic A. Hedgehog Pathway Activation Requires Coreceptor-Catalyzed, Lipid-Dependent Relay of the Sonic Hedgehog Ligand. Dev Cell 2020; 55:450-467.e8. [PMID: 33038332 DOI: 10.1016/j.devcel.2020.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/04/2020] [Accepted: 09/14/2020] [Indexed: 12/25/2022]
Abstract
Hedgehog signaling governs critical processes in embryogenesis, adult stem cell maintenance, and tumorigenesis. The activating ligand, Sonic hedgehog (SHH), is highly hydrophobic because of dual palmitate and cholesterol modification, and thus, its release from cells requires the secreted SCUBE proteins. We demonstrate that the soluble SCUBE-SHH complex, although highly potent in cellular assays, cannot directly signal through the SHH receptor, Patched1 (PTCH1). Rather, signaling by SCUBE-SHH requires a molecular relay mediated by the coreceptors CDON/BOC and GAS1, which relieves SHH inhibition by SCUBE. CDON/BOC bind both SCUBE and SHH, recruiting the complex to the cell surface. SHH is then handed off, in a dual lipid-dependent manner, to GAS1, and from GAS1 to PTCH1, initiating signaling. These results define an essential step in Hedgehog signaling, whereby coreceptors activate SHH by chaperoning it from a latent extracellular complex to its cell-surface receptor, and point to a broader paradigm of coreceptor function.
Collapse
Affiliation(s)
| | - Kostadin Petrov
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Laura Aravena
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Garrick Gu
- Williams College, Williamstown, MA 01267, USA
| | - Yangqing Xu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Ali H. SCUBE2, vascular endothelium, and vascular complications: A systematic review. Biomed Pharmacother 2020; 127:110129. [PMID: 32278240 DOI: 10.1016/j.biopha.2020.110129] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/11/2020] [Accepted: 03/25/2020] [Indexed: 10/24/2022] Open
Abstract
The vascular endothelium plays a vital role in regulating normal vascular function. Endothelial lining maintains the balance of thrombolytic and fibrinolytic microenvironment in the vasculature. Alterations of vascular endothelium referred to as endothelial dysfunction, caused the pathological changes in vessel wall such activation of proinflammatory and procoagulatory that initiate atherosclerosis. The concept that endothelial dysfunction plays a critical role in the initiation of atherosclerosis due to vascular inflammation gained tremendous attention. Diabetes mellitus is a metabolic-related disease that caused high mortality and morbidity, leading to its cardiovascular complication over the past decade. Atherosclerosis is the leading cardiovascular complication in diabetes mellitus. Despite metabolic and glycemic control, atherosclerotic plaque progression remains an enormous problem in diabetes mellitus complications. Thus, new inroads therapeutic approach in preventing complications that induced inflammation in endothelial cells could help prevent the disease progression. Signal peptide-CUB-EGF like domain-containing protein 2 (SCUBE2) expressed in vascular endothelium and reported to involve in inflammation. A recent study reported an increased SCUBE2 expression in diabetes mellitus and correlated with high expression of endothelin-1 (ET-1), a proinflammatory endothelial cell-derived peptide. Moreover, this gene showed to increase during atherosclerosis development. The present systematic review will summarize the involvement of SCUBE2 in vascular endothelium function changes and vascular complication, particularly in diabetes mellitus and atherosclerosis.
Collapse
Affiliation(s)
- Hirowati Ali
- Department of Biochemistry, Faculty of Medicine, Andalas University, Indonesia; Graduate School of Biomedical Sciences, Andalas University, Indonesia.
| |
Collapse
|
22
|
The Roles of Indian Hedgehog Signaling in TMJ Formation. Int J Mol Sci 2019; 20:ijms20246300. [PMID: 31847127 PMCID: PMC6941023 DOI: 10.3390/ijms20246300] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 12/10/2019] [Indexed: 01/15/2023] Open
Abstract
The temporomandibular joint (TMJ) is an intricate structure composed of the mandibular condyle, articular disc, and glenoid fossa in the temporal bone. Apical condylar cartilage is classified as a secondary cartilage, is fibrocartilaginous in nature, and is structurally distinct from growth plate and articular cartilage in long bones. Condylar cartilage is organized in distinct cellular layers that include a superficial layer that produces lubricants, a polymorphic/progenitor layer that contains stem/progenitor cells, and underlying layers of flattened and hypertrophic chondrocytes. Uniquely, progenitor cells reside near the articular surface, proliferate, undergo chondrogenesis, and mature into hypertrophic chondrocytes. During the past decades, there has been a growing interest in the molecular mechanisms by which the TMJ develops and acquires its unique structural and functional features. Indian hedgehog (Ihh), which regulates skeletal development including synovial joint formation, also plays pivotal roles in TMJ development and postnatal maintenance. This review provides a description of the many important recent advances in Hedgehog (Hh) signaling in TMJ biology. These include studies that used conventional approaches and those that analyzed the phenotype of tissue-specific mouse mutants lacking Ihh or associated molecules. The recent advances in understanding the molecular mechanism regulating TMJ development are impressive and these findings will have major implications for future translational medicine tools to repair and regenerate TMJ congenital anomalies and acquired diseases, such as degenerative damage in TMJ osteoarthritic conditions.
Collapse
|
23
|
Kim A, Savary C, Dubourg C, Carré W, Mouden C, Hamdi-Rozé H, Guyodo H, Douce JL, Pasquier L, Flori E, Gonzales M, Bénéteau C, Boute O, Attié-Bitach T, Roume J, Goujon L, Akloul L, Odent S, Watrin E, Dupé V, de Tayrac M, David V. Integrated clinical and omics approach to rare diseases: novel genes and oligogenic inheritance in holoprosencephaly. Brain 2019; 142:35-49. [PMID: 30508070 DOI: 10.1093/brain/awy290] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022] Open
Abstract
Holoprosencephaly is a pathology of forebrain development characterized by high phenotypic heterogeneity. The disease presents with various clinical manifestations at the cerebral or facial levels. Several genes have been implicated in holoprosencephaly but its genetic basis remains unclear: different transmission patterns have been described including autosomal dominant, recessive and digenic inheritance. Conventional molecular testing approaches result in a very low diagnostic yield and most cases remain unsolved. In our study, we address the possibility that genetically unsolved cases of holoprosencephaly present an oligogenic origin and result from combined inherited mutations in several genes. Twenty-six unrelated families, for whom no genetic cause of holoprosencephaly could be identified in clinical settings [whole exome sequencing and comparative genomic hybridization (CGH)-array analyses], were reanalysed under the hypothesis of oligogenic inheritance. Standard variant analysis was improved with a gene prioritization strategy based on clinical ontologies and gene co-expression networks. Clinical phenotyping and exploration of cross-species similarities were further performed on a family-by-family basis. Statistical validation was performed on 248 ancestrally similar control trios provided by the Genome of the Netherlands project and on 574 ancestrally matched controls provided by the French Exome Project. Variants of clinical interest were identified in 180 genes significantly associated with key pathways of forebrain development including sonic hedgehog (SHH) and primary cilia. Oligogenic events were observed in 10 families and involved both known and novel holoprosencephaly genes including recurrently mutated FAT1, NDST1, COL2A1 and SCUBE2. The incidence of oligogenic combinations was significantly higher in holoprosencephaly patients compared to two control populations (P < 10-9). We also show that depending on the affected genes, patients present with particular clinical features. This study reports novel disease genes and supports oligogenicity as clinically relevant model in holoprosencephaly. It also highlights key roles of SHH signalling and primary cilia in forebrain development. We hypothesize that distinction between different clinical manifestations of holoprosencephaly lies in the degree of overall functional impact on SHH signalling. Finally, we underline that integrating clinical phenotyping in genetic studies is a powerful tool to specify the clinical relevance of certain mutations.
Collapse
Affiliation(s)
- Artem Kim
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Clara Savary
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Christèle Dubourg
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Wilfrid Carré
- Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Charlotte Mouden
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Houda Hamdi-Rozé
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Hélène Guyodo
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Jerome Le Douce
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | | | | | | | - Elisabeth Flori
- Laboratoire de Cytogénétique, Cytologie et Histologie Quantitative, Hôpital de Hautepierre, HUS, Strasbourg, France
| | - Marie Gonzales
- Service de Génétique et Embryologie Médicales, Hôpital Armand Trousseau, Paris, France
| | | | | | - Tania Attié-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants-Malades, Université Paris Descartes, 149, rue de Sèvres, Paris, France
| | - Joelle Roume
- Department of Clinical Genetics, Centre de Référence “AnDDI Rares”, Poissy Hospital GHU PIFO, Poissy, France
| | | | - Linda Akloul
- Service de Génétique Clinique, CHU, Rennes, France
| | - Sylvie Odent
- Service de Génétique Clinique, CHU, Rennes, France
| | - Erwan Watrin
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Valérie Dupé
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France
| | - Marie de Tayrac
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| | - Véronique David
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000 Rennes, France.,Service de Génétique Moléculaire et Génomique, CHU, Rennes, France
| |
Collapse
|
24
|
Guo W, Roelink H. Loss of the Heparan Sulfate Proteoglycan Glypican5 Facilitates Long-Range Sonic Hedgehog Signaling. Stem Cells 2019; 37:899-909. [PMID: 30977233 PMCID: PMC8491322 DOI: 10.1002/stem.3018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 01/01/2023]
Abstract
As a morphogen, Sonic Hedgehog (Shh) mediates signaling at a distance from its sites of synthesis. After secretion, Shh must traverse a distance through the extracellular matrix (ECM) to reach the target cells and activate the Hh response. ECM proteins, in particular, the heparan sulfate proteoglycans (HSPGs) of the glypican family, have both negative and positive effects on Shh signaling, all attributed to their ability to bind Shh. Using mouse embryonic stem cell-derived mosaic tissues with compartments that lack the glycosyltransferases Exostosin1 and Exostosin2, or the HSPG core protein Glypican5, we show that Shh accumulates around its source cells when they are surrounded by cells that have a mutated ECM. This accumulation of Shh is correlated with an increased noncell autonomous Shh response. Our results support a model in which Shh presented on the cell surface accumulates at or near ECM that lacks HSPGs, possibly due to the absence of these Shh sequestering molecules. Stem Cells 2019;37:899-909.
Collapse
Affiliation(s)
- Wei Guo
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California, USA
| | - Henk Roelink
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, California, USA
| |
Collapse
|
25
|
Manikowski D, Jakobs P, Jboor H, Grobe K. Soluble Heparin and Heparan Sulfate Glycosaminoglycans Interfere with Sonic Hedgehog Solubilization and Receptor Binding. Molecules 2019; 24:molecules24081607. [PMID: 31018591 PMCID: PMC6526471 DOI: 10.3390/molecules24081607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/24/2022] Open
Abstract
Sonic hedgehog (Shh) signaling plays a tumor-promoting role in many epithelial cancers. Cancer cells produce soluble a Shh that signals to distant stromal cells that express the receptor Patched (Ptc). These receiving cells respond by producing other soluble factors that promote cancer cell growth, generating a positive feedback loop. To interfere with reinforced Shh signaling, we examined the potential of defined heparin and heparan sulfate (HS) polysaccharides to block Shh solubilization and Ptc receptor binding. We confirm in vitro and in vivo that proteolytic cleavage of the N-terminal Cardin-Weintraub (CW) amino acid motif is a prerequisite for Shh solubilization and function. Consistent with the established binding of soluble heparin or HS to the Shh CW target motif, both polysaccharides impaired proteolytic Shh processing and release from source cells. We also show that HS and heparin bind to, and block, another set of basic amino acids required for unimpaired Shh binding to Ptc receptors on receiving cells. Both modes of Shh activity downregulation depend more on HS size and overall charge than on specific HS sulfation modifications. We conclude that heparin oligosaccharide interference in the physiological roles of HS in Shh release and reception may be used to expand the field of investigation to pharmaceutical intervention of tumor-promoting Shh functions.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- Binding, Competitive
- Cell Line, Tumor
- Drosophila Proteins/antagonists & inhibitors
- Drosophila Proteins/chemistry
- Drosophila Proteins/genetics
- Drosophila Proteins/metabolism
- Drosophila melanogaster/genetics
- Drosophila melanogaster/growth & development
- Drosophila melanogaster/metabolism
- Embryo, Nonmammalian
- Feedback, Physiological
- Gene Expression Regulation, Developmental
- HeLa Cells
- Hedgehog Proteins/antagonists & inhibitors
- Hedgehog Proteins/chemistry
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Heparin/chemistry
- Heparin/pharmacology
- Heparitin Sulfate/chemistry
- Heparitin Sulfate/pharmacology
- Humans
- Models, Molecular
- Patched-1 Receptor/genetics
- Patched-1 Receptor/metabolism
- Protein Binding
- Protein Interaction Domains and Motifs
- Protein Structure, Secondary
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction
- Solubility
- Wings, Animal/growth & development
- Wings, Animal/metabolism
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany.
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany.
| | - Hamodah Jboor
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany.
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
26
|
Casillas C, Roelink H. Gain-of-function Shh mutants activate Smo cell-autonomously independent of Ptch1/2 function. Mech Dev 2018; 153:30-41. [PMID: 30144507 PMCID: PMC6165682 DOI: 10.1016/j.mod.2018.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
Sonic Hedgehog (Shh) signaling is characterized by non-cell autonomy; cells expressing Shh do not respond to the ligand. Here, we identify several Shh mutations that can activate the Hedgehog (Hh) pathway cell-autonomously. Cell-autonomous pathway activation requires the extracellular cysteine rich domain of Smoothened, but is otherwise independent of the Shh receptors Patched1 and -2. Many of the Shh mutants that gain activity fail to undergo auto processing resulting in the perdurance of the Shh pro-peptide, a form of Shh that is sufficient to activate the Hh response cell-autonomously. Our results demonstrate that Shh is capable of activating the Hh pathway via Smoothened, independently of Patched1/2, and that it harbors an intrinsic mechanism that prevents cell-autonomous activation of the Shh response.
Collapse
Affiliation(s)
- Catalina Casillas
- Department of Molecular and Cell Biology, 16 Barker Hall, 3204, University of California, Berkeley, CA 94720, USA
| | - Henk Roelink
- Department of Molecular and Cell Biology, 16 Barker Hall, 3204, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
27
|
Kastl P, Manikowski D, Steffes G, Schürmann S, Bandari S, Klämbt C, Grobe K. Disrupting Hedgehog Cardin-Weintraub sequence and positioning changes cellular differentiation and compartmentalization in vivo. Development 2018; 145:145/18/dev167221. [PMID: 30242104 DOI: 10.1242/dev.167221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022]
Abstract
Metazoan Hedgehog (Hh) morphogens are essential regulators of growth and patterning at significant distances from their source, despite being produced as N-terminally palmitoylated and C-terminally cholesteroylated proteins, which firmly tethers them to the outer plasma membrane leaflet of producing cells and limits their spread. One mechanism to overcome this limitation is proteolytic processing of both lipidated terminal peptides, called shedding, but molecular target site requirements for effective Hh shedding remained undefined. In this work, by using Drosophila melanogaster as a model, we show that mutagenesis of the N-terminal Cardin-Weintraub (CW) motif inactivates recombinant Hh proteins to variable degrees and, if overexpressed in the same compartment, converts them into suppressors of endogenous Hh function. In vivo, additional removal of N-palmitate membrane anchors largely restored endogenous Hh function, supporting the hypothesis that proteolytic CW processing controls Hh solubilization. Importantly, we also observed that CW repositioning impairs anterior/posterior compartmental boundary maintenance in the third instar wing disc. This demonstrates that Hh shedding not only controls the differentiation of anterior cells, but also maintains the sharp physical segregation between these receiving cells and posterior Hh-producing cells.
Collapse
Affiliation(s)
- Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Georg Steffes
- Institute of Neurobiology and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Shyam Bandari
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Christian Klämbt
- Institute of Neurobiology and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
28
|
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
|
29
|
Schürmann S, Steffes G, Manikowski D, Kastl P, Malkus U, Bandari S, Ohlig S, Ortmann C, Rebollido-Rios R, Otto M, Nüsse H, Hoffmann D, Klämbt C, Galic M, Klingauf J, Grobe K. Proteolytic processing of palmitoylated Hedgehog peptides specifies the 3-4 intervein region of the Drosophila wing. eLife 2018. [PMID: 29522397 PMCID: PMC5844694 DOI: 10.7554/elife.33033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell fate determination during development often requires morphogen transport from producing to distant responding cells. Hedgehog (Hh) morphogens present a challenge to this concept, as all Hhs are synthesized as terminally lipidated molecules that form insoluble clusters at the surface of producing cells. While several proposed Hh transport modes tie directly into these unusual properties, the crucial step of Hh relay from producing cells to receptors on remote responding cells remains unresolved. Using wing development in Drosophila melanogaster as a model, we show that Hh relay and direct patterning of the 3–4 intervein region strictly depend on proteolytic removal of lipidated N-terminal membrane anchors. Site-directed modification of the N-terminal Hh processing site selectively eliminated the entire 3–4 intervein region, and additional targeted removal of N-palmitate restored its formation. Hence, palmitoylated membrane anchors restrict morphogen spread until site-specific processing switches membrane-bound Hh into bioactive forms with specific patterning functions. Each cell in a developing embryo receives information that determines what type of body structure it will form. In fruit flies, this information is partly given by a protein called Hedgehog. In the embryo cells that receive it, Hedgehog can trigger a series of events which activate certain genes and thereby regulate structure formation. The Hedgehog proteins are produced by a different organizing group of cells: from there they transport within the embryo, creating a gradient. Depending on where a responding cell is in the embryo, it receives a different amount of Hedgehog, which gives the cell its identity. For example, Hedgehog proteins form a gradient across a fruit fly’s developing wing, which creates a visible vein pattern. How Hedgehog proteins form gradients is enigmatic, however, because once produced, they cling to the cells that created them. The reason for this unusual behavior is that the two ends of the Hedgehog protein are attached to a different fat molecule. In particular, one extremity is linked to a fat molecule called palmitate. These ends’ fatty additions anchor Hedgehog to the cells that produced them. Then, the tethered proteins gather together to form chain-like clusters where they inactivate each other: the extremity with the palmitate ‘hides’ the portion of the neighboring protein that binds to the receiving cells. It is still unclear how Hedgehog can be activated and released to reach these faraway cells. One hypothesis is that an enzyme comes to the clusters and frees the proteins by cutting both of Hedgehog’s fatty anchors. Thanks to how the palmitate tethers Hedgehog to the cell, the protein is positioned in such a way that when the enzyme makes its snip, the binding site on the neighboring Hedgehog gets exposed: this protein is activated and, when also cut by the enzyme, released. Here, Schürmann et al. create an array of mutant Hedgehog proteins – for example some without palmitate, some with palmitate that cannot be removed by the enzyme – and study how they affect the development of the wing’s pattern in the fruit fly. Coupled with the imaging of the clusters, these experiments support the hypothesis that the palmitate anchor is necessary so that Hedgehog proteins can be turned on before diffusing away. The Hedgehog family of proteins is also present in humans, where it presides over the development of the embryo but is also involved in cancer. Understanding how Hedgehog works in the fruit fly could lead to new discoveries in humans too.
Collapse
Affiliation(s)
- Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Georg Steffes
- Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.,Institute of Neurobiology, University of Münster, Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Ursula Malkus
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Shyam Bandari
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Stefanie Ohlig
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Corinna Ortmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | | | - Mandy Otto
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Harald Nüsse
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Daniel Hoffmann
- Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Christian Klämbt
- Institute of Neurobiology, University of Münster, Münster, Germany
| | - Milos Galic
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
30
|
Manikowski D, Kastl P, Grobe K. Taking the Occam's Razor Approach to Hedgehog Lipidation and Its Role in Development. J Dev Biol 2018; 6:jdb6010003. [PMID: 29615552 PMCID: PMC5875562 DOI: 10.3390/jdb6010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 01/01/2023] Open
Abstract
All Hedgehog (Hh) proteins signal from producing cells to distant receiving cells despite being synthesized as N-and C-terminally lipidated, membrane-tethered molecules. To explain this paradoxical situation, over the past 15 years, several hypotheses have been postulated that tie directly into this property, such as Hh transport on cellular extensions called cytonemes or on secreted vesicles called lipophorins and exosomes. The alternative situation that tight membrane association merely serves to prevent unregulated Hh solubilization has been addressed by biochemical and structural studies suggesting Hh extraction from the membrane or proteolytic Hh release. While some of these models may act in different organisms, tissues or developmental programs, others may act together to specify Hh short- and long-range signaling in the same tissues. To test and rank these possibilities, we here review major models of Hh release and transport and hypothesize that the (bio)chemical and physical properties of firmly established, homologous, and functionally essential biochemical Hh modifications are adapted to specify and determine interdependent steps of Hh release, transport and signaling, while ruling out other steps. This is also described by the term “congruence”, meaning that the logical combination of biochemical Hh modifications can reveal their true functional implications. This combined approach reveals potential links between models of Hh release and transport that were previously regarded as unrelated, thereby expanding our view of how Hhs can steer development in a simple, yet extremely versatile, manner.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
31
|
Matrix metalloproteinase-9 activity and a downregulated Hedgehog pathway impair blood-brain barrier function in an in vitro model of CNS tuberculosis. Sci Rep 2017; 7:16031. [PMID: 29167512 PMCID: PMC5700087 DOI: 10.1038/s41598-017-16250-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/03/2017] [Indexed: 12/21/2022] Open
Abstract
Central nervous system tuberculosis (CNS TB) has a high mortality and morbidity associated with severe inflammation. The blood-brain barrier (BBB) protects the brain from inflammation but the mechanisms causing BBB damage in CNS TB are uncharacterized. We demonstrate that Mycobacterium tuberculosis (Mtb) causes breakdown of type IV collagen and decreases tight junction protein (TJP) expression in a co-culture model of the BBB. This increases permeability, surface expression of endothelial adhesion molecules and leukocyte transmigration. TJP breakdown was driven by Mtb-dependent secretion of matrix metalloproteinase (MMP)-9. TJP expression is regulated by Sonic hedgehog (Shh) through transcription factor Gli-1. In our model, the hedgehog pathway was downregulated by Mtb-stimulation, but Shh levels in astrocytes were unchanged. However, Scube2, a glycoprotein regulating astrocyte Shh release was decreased, inhibiting Shh delivery to brain endothelial cells. Activation of the hedgehog pathway by addition of a Smoothened agonist or by addition of exogenous Shh, or neutralizing MMP-9 activity, decreased permeability and increased TJP expression in the Mtb-stimulated BBB co-cultures. In summary, the BBB is disrupted by downregulation of the Shh pathway and breakdown of TJPs, secondary to increased MMP-9 activity which suggests that these pathways are potential novel targets for host directed therapy in CNS TB.
Collapse
|
32
|
Jakobs P, Schulz P, Schürmann S, Niland S, Exner S, Rebollido-Rios R, Manikowski D, Hoffmann D, Seidler DG, Grobe K. Ca 2+ coordination controls sonic hedgehog structure and its Scube2-regulated release. J Cell Sci 2017; 130:3261-3271. [PMID: 28778988 DOI: 10.1242/jcs.205872] [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: 05/04/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Proteolytic processing of cell-surface-bound ligands, called shedding, is a fundamental system to control cell-cell signaling. Yet, our understanding of how shedding is regulated is still incomplete. One way to increase the processing of dual-lipidated membrane-associated Sonic hedgehog (Shh) is to increase the density of substrate and sheddase. This releases and also activates Shh by the removal of lipidated inhibitory N-terminal peptides from Shh receptor binding sites. Shh release and activation is enhanced by Scube2 [signal sequence, cubulin (CUB) domain, epidermal growth factor (EGF)-like protein 2], raising the question of how this is achieved. Here, we show that Scube2 EGF domains are responsible for specific proteolysis of the inhibitory Shh N-terminus, and that CUB domains complete the process by reversing steric masking of this peptide. Steric masking, in turn, depends on Ca2+ occupancy of Shh ectodomains, unveiling a new mode of shedding regulation at the substrate level. Importantly, Scube2 uncouples processing of Shh peptides from their lipid-mediated juxtamembrane positioning, and thereby explains the long-standing conundrum that N-terminally unlipidated Shh shows patterning activity in Scube2-expressing vertebrates, but not in invertebrates that lack Scube orthologs.
Collapse
Affiliation(s)
- Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Philipp Schulz
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Sebastian Exner
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Rocio Rebollido-Rios
- Center for Medical Biotechnology, University of Duisburg-Essen, D-45117 Essen, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Daniel Hoffmann
- Center for Medical Biotechnology, University of Duisburg-Essen, D-45117 Essen, Germany
| | - Daniela G Seidler
- Centre for Internal Medicine, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School I3, EB2/R3110, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
33
|
Shilo BZ, Barkai N. Buffering Global Variability of Morphogen Gradients. Dev Cell 2017; 40:429-438. [DOI: 10.1016/j.devcel.2016.12.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/14/2016] [Accepted: 12/05/2016] [Indexed: 12/23/2022]
|
34
|
Simon E, Aguirre-Tamaral A, Aguilar G, Guerrero I. Perspectives on Intra- and Intercellular Trafficking of Hedgehog for Tissue Patterning. J Dev Biol 2016; 4:jdb4040034. [PMID: 29615597 PMCID: PMC5831803 DOI: 10.3390/jdb4040034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/14/2022] Open
Abstract
Intercellular communication is a fundamental process for correct tissue development. The mechanism of this process involves, among other things, the production and secretion of signaling molecules by specialized cell types and the capability of these signals to reach the target cells in order to trigger specific responses. Hedgehog (Hh) is one of the best-studied signaling pathways because of its importance during morphogenesis in many organisms. The Hh protein acts as a morphogen, activating its targets at a distance in a concentration-dependent manner. Post-translational modifications of Hh lead to a molecule covalently bond to two lipid moieties. These lipid modifications confer Hh high affinity to lipidic membranes, and intense studies have been carried out to explain its release into the extracellular matrix. This work reviews Hh molecule maturation, the intracellular recycling needed for its secretion and the proposed carriers to explain Hh transportation to the receiving cells. Special focus is placed on the role of specialized filopodia, also named cytonemes, in morphogen transport and gradient formation.
Collapse
Affiliation(s)
- Eléanor Simon
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Adrián Aguirre-Tamaral
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Gustavo Aguilar
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Isabel Guerrero
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
35
|
Guo E, Liu H, Liu X. Overexpression of SCUBE2 Inhibits Proliferation, Migration, and Invasion in Glioma Cells. Oncol Res 2016; 25:437-444. [PMID: 27697090 PMCID: PMC7841219 DOI: 10.3727/096504016x14747335734344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Signal peptide CUB EGF-like domain-containing protein 2 (SCUBE2), a member of the SCUBE family of proteins, was recently found to play an important role in cancer development. However, little is known regarding its biological function in glioma. In the present study, we investigated the effect of SCUBE2 on glioma and explored its relevant mechanisms. The study showed that SCUBE2 had a low expression in glioma tissue and cell lines. SCUBE2 overexpression inhibited glioma cell proliferation in vitro and in vivo as well as suppressed glioma cell migration and invasion in vitro. Furthermore, we found that the Sonic hedgehog (Shh) signaling pathway was involved in the inhibitory effect of SCUBE2 overexpression on glioma cells. In light of the results obtained from our study, SCUBE2 may be regarded as a potential therapeutic target for glioma.
Collapse
|
36
|
Ramsbottom SA, Pownall ME, Roelink H, Conway SJ. Regulation of Hedgehog Signalling Inside and Outside the Cell. J Dev Biol 2016; 4:23. [PMID: 27547735 PMCID: PMC4990124 DOI: 10.3390/jdb4030023] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The hedgehog (Hh) signalling pathway is conserved throughout metazoans and plays an important regulatory role in both embryonic development and adult homeostasis. Many levels of regulation exist that control the release, reception, and interpretation of the hedgehog signal. The fatty nature of the Shh ligand means that it tends to associate tightly with the cell membrane, and yet it is known to act as a morphogen that diffuses to elicit pattern formation. Heparan sulfate proteoglycans (HSPGs) play a major role in the regulation of Hh distribution outside the cell. Inside the cell, the primary cilium provides an important hub for processing the Hh signal in vertebrates. This review will summarise the current understanding of how the Hh pathway is regulated from ligand production, release, and diffusion, through to signal reception and intracellular transduction.
Collapse
Affiliation(s)
- Simon A. Ramsbottom
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, NE1 3BZ Newcastle upon Tyne, UK
- Correspondence: ; Tel.: +44-(0)191-241-8612
| | | | | | | |
Collapse
|
37
|
Abstract
The Hedgehog (Hh) signalling pathway is one of the key regulators of metazoan development. Hh proteins have been shown to play roles in many developmental processes and have become paradigms for classical morphogens. Dysfunction of the Hh pathway underlies a number of human developmental abnormalities and diseases, making it an important therapeutic target. Interest in Hh signalling thus extends across many fields, from evo-devo to cancer research and regenerative medicine. Here, and in the accompanying poster, we provide an outline of the current understanding of Hh signalling mechanisms, highlighting the similarities and differences between species.
Collapse
Affiliation(s)
- Raymond Teck Ho Lee
- Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A* STAR), Singapore, 138673 Singapore
| | - Zhonghua Zhao
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921 Singapore
| | - Philip W Ingham
- Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A* STAR), Singapore, 138673 Singapore Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921 Singapore Department of Medicine, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
38
|
Resh MD. Fatty acylation of proteins: The long and the short of it. Prog Lipid Res 2016; 63:120-31. [PMID: 27233110 DOI: 10.1016/j.plipres.2016.05.002] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/19/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022]
Abstract
Long, short and medium chain fatty acids are covalently attached to hundreds of proteins. Each fatty acid confers distinct biochemical properties, enabling fatty acylation to regulate intracellular trafficking, subcellular localization, protein-protein and protein-lipid interactions. Myristate and palmitate represent the most common fatty acid modifying groups. New insights into how fatty acylation reactions are catalyzed, and how fatty acylation regulates protein structure and function continue to emerge. Myristate is typically linked to an N-terminal glycine, but recent studies reveal that lysines can also be myristoylated. Enzymes that remove N-terminal myristoyl-glycine or myristate from lysines have now been identified. DHHC proteins catalyze S-palmitoylation, but the mechanisms that regulate substrate recognition by individual DHHC family members remain to be determined. New studies continue to reveal thioesterases that remove palmitate from S-acylated proteins. Another area of rapid expansion is fatty acylation of the secreted proteins hedgehog, Wnt and Ghrelin, by Hhat, Porcupine and GOAT, respectively. Understanding how these membrane bound O-acyl transferases recognize their protein and fatty acyl CoA substrates is an active area of investigation, and is punctuated by the finding that these enzymes are potential drug targets in human diseases.
Collapse
Affiliation(s)
- Marilyn D Resh
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 143, New York, NY 10075, United States.
| |
Collapse
|
39
|
Jakobs P, Schulz P, Ortmann C, Schürmann S, Exner S, Rebollido-Rios R, Dreier R, Seidler DG, Grobe K. Bridging the gap: heparan sulfate and Scube2 assemble Sonic hedgehog release complexes at the surface of producing cells. Sci Rep 2016; 6:26435. [PMID: 27199253 PMCID: PMC4873810 DOI: 10.1038/srep26435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022] Open
Abstract
Decision making in cellular ensembles requires the dynamic release of signaling molecules from the producing cells into the extracellular compartment. One important example of molecules that require regulated release in order to signal over several cell diameters is the Hedgehog (Hh) family, because all Hhs are synthesized as dual-lipidated proteins that firmly tether to the outer membrane leaflet of the cell that produces them. Factors for the release of the vertebrate Hh family member Sonic Hedgehog (Shh) include cell-surface sheddases that remove the lipidated terminal peptides, as well as the soluble glycoprotein Scube2 that cell-nonautonomously enhances this process. This raises the question of how soluble Scube2 is recruited to cell-bound Shh substrates to regulate their turnover. We hypothesized that heparan sulfate (HS) proteoglycans (HSPGs) on the producing cell surface may play this role. In this work, we confirm that HSPGs enrich Scube2 at the surface of Shh-producing cells and that Scube2-regulated proteolytic Shh processing and release depends on specific HS. This finding indicates that HSPGs act as cell-surface assembly and storage platforms for Shh substrates and for protein factors required for their release, making HSPGs critical decision makers for Scube2-dependent Shh signaling from the surface of producing cells.
Collapse
Affiliation(s)
- P Jakobs
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - P Schulz
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - C Ortmann
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - S Schürmann
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - S Exner
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - R Rebollido-Rios
- Center for Medical Biotechnology#, University of Duisburg-Essen, 45117 Essen, Germany
| | - R Dreier
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - D G Seidler
- Centre for Internal Medicine, Hannover Medical School I3, EB2/R3110, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - K Grobe
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| |
Collapse
|
40
|
Gurdziel K, Vogt KR, Schneider G, Richards N, Gumucio DL. Computational prediction and experimental validation of novel Hedgehog-responsive enhancers linked to genes of the Hedgehog pathway. BMC DEVELOPMENTAL BIOLOGY 2016; 16:4. [PMID: 26912062 PMCID: PMC4765071 DOI: 10.1186/s12861-016-0106-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/16/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND The Hedgehog (Hh) signaling pathway, acting through three homologous transcription factors (GLI1, GLI2, GLI3) in vertebrates, plays multiple roles in embryonic organ development and adult tissue homeostasis. At the level of the genome, GLI factors bind to specific motifs in enhancers, some of which are hundreds of kilobases removed from the gene promoter. These enhancers integrate the Hh signal in a context-specific manner to control the spatiotemporal pattern of target gene expression. Importantly, a number of genes that encode Hh pathway molecules are themselves targets of Hh signaling, allowing pathway regulation by an intricate balance of feed-back activation and inhibition. However, surprisingly few of the critical enhancer elements that control these pathway target genes have been identified despite the fact that such elements are central determinants of Hh signaling activity. Recently, ChIP studies have been carried out in multiple tissue contexts using mouse models carrying FLAG-tagged GLI proteins (GLI(FLAG)). Using these datasets, we tested whether a meta-analysis of GLI binding sites, coupled with a machine learning approach, could reveal genomic features that could be used to empirically identify Hh-regulated enhancers linked to loci of the Hh signaling pathway. RESULTS A meta-analysis of four existing GLI(FLAG) datasets revealed a library of GLI binding motifs that was substantially more restricted than the potential sites predicted by previous in vitro binding studies. A machine learning method (kmer-SVM) was then applied to these datasets and enriched k-mers were identified that, when applied to the mouse genome, predicted as many as 37,000 potential Hh enhancers. For functional analysis, we selected nine regions which were annotated to putative Hh pathway molecules and found that seven exhibited GLI-dependent activity, indicating that they are directly regulated by Hh signaling (78% success rate). CONCLUSIONS The results suggest that Hh enhancer regions share common sequence features. The kmer-SVM machine learning approach identifies those features and can successfully predict functional Hh regulatory regions in genomic DNA surrounding Hh pathway molecules and likely, other Hh targets. Additionally, the library of enriched GLI binding motifs that we have identified may allow improved identification of functional GLI binding sites.
Collapse
Affiliation(s)
- Katherine Gurdziel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kyle R Vogt
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gary Schneider
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Neil Richards
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
41
|
Not so Fast: Co-Requirements for Sonic Hedgehog Induced Brain Tumorigenesis. Cancers (Basel) 2015; 7:1484-98. [PMID: 26258793 PMCID: PMC4586781 DOI: 10.3390/cancers7030848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 01/17/2023] Open
Abstract
The Sonic hedgehog (Shh) pathway plays an integral role in cellular proliferation during normal brain development and also drives growth in a variety of cancers including brain cancer. Clinical trials of Shh pathway inhibitors for brain tumors have yielded disappointing results, indicating a more nuanced role for Shh signaling. We postulate that Shh signaling does not work alone but requires co-activation of other signaling pathways for tumorigenesis and stem cell maintenance. This review will focus on the interplay between the Shh pathway and these pathways to promote tumor growth in brain tumors, presenting opportunities for the study of combinatorial therapies.
Collapse
|
42
|
Lin YC, Roffler SR, Yan YT, Yang RB. Disruption of Scube2 Impairs Endochondral Bone Formation. J Bone Miner Res 2015; 30:1255-67. [PMID: 25639508 DOI: 10.1002/jbmr.2451] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 11/11/2022]
Abstract
Signal peptide-CUB-EGF domain-containing protein 2 (SCUBE2) belongs to a secreted and membrane-tethered multidomain SCUBE protein family composed of three members found in vertebrates and mammals. Recent reports suggested that zebrafish scube2 could facilitate sonic hedgehog (Shh) signaling for proper development of slow muscle. However, whether SCUBE2 can regulate the signaling activity of two other hedgehog ligands (Ihh and Dhh), and the developmental relevance of the SCUBE2-induced hedgehog signaling in mammals remain poorly understood. In this study, we first showed that as compared with SCUBE1 or SCUBE3, SCUBE2 is the most potent modulator of IHH signaling in vitro. In addition, gain and loss-of-function studies demonstrated that SCUBE2 exerted an osteogenic function by enhancing Ihh-stimulated osteoblast differentiation in the mouse mesenchymal progenitor cells. Consistent with these in vitro studies and the prominent roles of Ihh in coordinating skeletogenesis, genetic ablation of Scube2 (-/-) caused defective endochondral bone formation and impaired Ihh-mediated chondrocyte differentiation and proliferation as well as osteoblast differentiation of -/- bone-marrow mesenchymal stromal-cell cultures. Our data demonstrate that Scube2 plays a key regulatory role in Ihh-dependent endochondral bone formation.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
43
|
Ortmann C, Pickhinke U, Exner S, Ohlig S, Lawrence R, Jboor H, Dreier R, Grobe K. Sonic hedgehog processing and release are regulated by glypican heparan sulfate proteoglycans. J Cell Sci 2015; 128:2374-85. [PMID: 25967551 DOI: 10.1242/jcs.170670] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022] Open
Abstract
All Hedgehog morphogens are released from producing cells, despite being synthesized as N- and C-terminally lipidated molecules, a modification that firmly tethers them to the cell membrane. We have previously shown that proteolytic removal of both lipidated peptides, called shedding, releases bioactive Sonic hedgehog (Shh) morphogens from the surface of transfected Bosc23 cells. Using in vivo knockdown together with in vitro cell culture studies, we now show that glypican heparan sulfate proteoglycans regulate this process, through their heparan sulfate chains, in a cell autonomous manner. Heparan sulfate specifically modifies Shh processing at the cell surface, and purified glycosaminoglycans enhance the proteolytic removal of N- and C-terminal Shh peptides under cell-free conditions. The most likely explanation for these observations is direct Shh processing in the extracellular compartment, suggesting that heparan sulfate acts as a scaffold or activator for Shh ligands and the factors required for their turnover. We also show that purified heparan sulfate isolated from specific cell types and tissues mediates the release of bioactive Shh from pancreatic cancer cells, revealing a previously unknown regulatory role for these versatile molecules in a pathological context.
Collapse
Affiliation(s)
- Corinna Ortmann
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Ute Pickhinke
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Sebastian Exner
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Stefanie Ohlig
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Roger Lawrence
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hamodah Jboor
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Rita Dreier
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Kay Grobe
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| |
Collapse
|
44
|
Abstract
Cholesterylation is a post-translational attachment of sterol to proteins. This modification has been a characteristic of a single family of hedgehog proteins (Hh). Hh is a well-established morphogenic molecule important in embryonic development. It was also found to be involved in the progression of many cancer types. Herein, we describe the mechanism of biosynthesis of cholesterylated Hh, the role of this unusual modification on protein functions and novel chemical probes, which could be used to specifically target this modification, both in vitro and in vivo.
Collapse
|