1
|
Hezwani M, Anokye D, Soutar DE, Ligorio M, Prabhakar N, Oram JC, Cantor AJ, Jackson GD, Terracciano R, Walker M, Baker AN. Glycopolymer-Functionalized Gold Nanoparticles for the Detection of Western Diamondback Rattlesnake ( Crotalus atrox) Venom. Biomacromolecules 2025. [PMID: 40392118 DOI: 10.1021/acs.biomac.5c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Every 5 minutes, 50 people are bitten by a snake worldwide; four will be permanently disabled and one will die. Most approaches to treating and diagnosing snake envenomation, a World Health Organization (WHO)-neglected tropical disease, rely on antibody-based solutions derived from animals or cell culture. Here, we present the first proof of concept for a glycopolymer-based ultraviolet-visible (UV-vis) assay to detect snake venom, specifically Western Diamondback Rattlesnake (Crotalus atrox) venom. This was achieved by synthesizing a library of glycan-terminated poly(hydroxyethyl acrylamide) functionalized gold nanoparticles. The library was analyzed using UV-vis spectroscopy and biolayer interferometry, with galactose-terminating systems found to demonstrate specificity for C. atrox venom, versus model lectins and Naja naja venom in UV-vis assays. This corroborates glycan array data in the literature and highlights our glycopolymer systems' potential as a diagnostic tool for snakebite, with the best particle system displaying a limit of detection of ∼20 μg·mL-1.
Collapse
Affiliation(s)
- Mahdi Hezwani
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Derecash Anokye
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Douglas E Soutar
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Melissa Ligorio
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Neil Prabhakar
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Jack C Oram
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Alexander J Cantor
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Garrett D Jackson
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Roberto Terracciano
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Marc Walker
- Department of Physics, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| | - Alexander N Baker
- Department of Chemistry, University of Warwick, Gibbet Hill Road, CV4 7AL Coventry, U.K
| |
Collapse
|
2
|
Woodward AM, Guindolet D, Martinez-Carrasco R, Gabison EE, Lavker RM, Argüeso P. Low fucosylation defines the glycocalyx of progenitor cells and melanocytes in the human limbal stem cell niche. Stem Cell Reports 2025; 20:102378. [PMID: 39706176 PMCID: PMC11784483 DOI: 10.1016/j.stemcr.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024] Open
Abstract
It is widely recognized that the glycocalyx has significant implications in regulating the self-renewal and differentiation of adult stem cells; however, its composition remains poorly understood. Here, we show that the fucose-binding Aleuria aurantia lectin (AAL) binds differentially to basal cells in the stratified epithelium of the human limbus, hair follicle epithelium, and meibomian gland duct. Using fluorescence-activated cell sorting in combination with single-cell transcriptomics, we find that most epithelial progenitor cells and melanocytes in the limbus display low AAL staining (AALlow) on their cell surface, an attribute that is gradually lost in epithelial cells as they differentiate into mature corneal cells. AALlow epithelial cells were enriched in putative limbal stem cell markers and displayed high clonogenic capacity. Further analyses revealed that AALlow epithelial cells had reduced expression of GDP-mannose-4,6-dehydratase, an enzyme catalyzing the first and limiting step in the de novo biosynthesis of GDP-fucose, and that inhibition of fucosylation using a small-molecule fucose analog stimulated the proliferative potential of limbal epithelial cells ex vivo. These results provide crucial insights into the distinctive composition of the glycocalyx in adult stem cells and underscore the significance of fucose modulation in the therapeutic regeneration of the human limbal stem cell niche.
Collapse
Affiliation(s)
- Ashley M Woodward
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | - Damien Guindolet
- Fondation Ophtalmologique A. de Rothschild, 25 rue Manin, 75019 Paris, France
| | - Rafael Martinez-Carrasco
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | - Eric E Gabison
- Fondation Ophtalmologique A. de Rothschild, 25 rue Manin, 75019 Paris, France
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Pablo Argüeso
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
3
|
Yu F, Lou S, He H, Zhou Y. Potential role of POFUT1 as a prognostic predictor in low-grade gliomas: Immune microenvironment insights from a pan-cancer analysis. Heliyon 2024; 10:e27004. [PMID: 38463813 PMCID: PMC10923674 DOI: 10.1016/j.heliyon.2024.e27004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 01/31/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
The POFUT1 gene, known to be up-regulated in various tumor tissues and associated with tumor biology, has yet to be explored for its potential role in immune response regulation and tumor immune microenvironment. The normalized pan-cancer dataset (TCGA Pan-Cancer) was downloaded from the UCSC database, followed by analysis of POFUT1 expression in various tumors and functional enrichment analysis. The correlation between POFUT1 expression levels and patient prognosis was assessed. GSEA of POFUT1 based on low-grade glioma (LGG) samples and immune infiltration analyses of LGG and glioblastoma (GBM) were conducted. The correlation between POFUT1 expression levels and infiltration levels of 22 immune cells in LGG and GBM was examined, as well as the correlation between immune cell infiltration levels and LGG patient prognosis. Additionally, the relationship between POFUT1 expression levels and characteristic gene expression of identified immune cells was evaluated. Lastly, external dataset validation was performed using the integrated CGGA dataset. Significant differences were observed in POFUT1 expression levels across 20 tumor types. High POFUT1 expression correlated with poor prognosis in GBMLGG, and LGG patients. Enrichment analysis and GSEA of POFUT1 in LGG demonstrated involvement in tumor-related and immune-related pathways. A positive correlation was identified between POFUT1 expression levels and infiltration levels of resting memory CD4+ T cells, as well as M2 macrophages or M2-like TAMs in the LGG immune microenvironment, potentially contributing to poor prognosis. External dataset validation revealed a positive correlation between M2 macrophages or M2-like TAMs and POFUT1 expression levels in LGG, and a negative correlation with LGG patient prognosis. POFUT1's negative impact on LGG prognosis may result from its influence on M2 macrophage and M2-like TAM infiltration levels within the immune microenvironment. This suggests its potential as a prognostic predictor and therapeutic target for LGG.
Collapse
Affiliation(s)
- Fan Yu
- Centre of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| | - Shuang Lou
- Centre of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| | - Haihong He
- Centre of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| | - Yiwen Zhou
- Centre of Clinical Laboratory Medicine, Shenzhen Hospital, Southern Medical University, China
| |
Collapse
|
4
|
Stanley P, Tanwar A. Regulation of myeloid and lymphoid cell development by O-glycans on Notch. Front Mol Biosci 2022; 9:979724. [PMID: 36406268 PMCID: PMC9672378 DOI: 10.3389/fmolb.2022.979724] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/13/2022] [Indexed: 10/06/2023] Open
Abstract
Notch signaling via NOTCH1 stimulated by Delta-like ligand 4 (DLL4) is required for the development of T cells in thymus, and NOTCH2 stimulated by Notch ligand DLL1 is required for the development of marginal zone (MZ) B cells in spleen. Notch signaling also regulates myeloid cell production in bone marrow and is an essential contributor to the generation of early hematopoietic stem cells (HSC). The differentiation program in each of these cellular contexts is optimized by the regulation of Notch signaling strength by O-glycans attached to epidermal growth factor-like (EGF) repeats in the extracellular domain of Notch receptors. There are three major types of O-glycan on NOTCH1 and NOTCH2 - O-fucose, O-glucose and O-GlcNAc. The initiating sugar of each O-glycan is added in the endoplasmic reticulum (ER) by glycosyltransferases POFUT1 (fucose), POGLUT1/2/3 (glucose) or EOGT (GlcNAc), respectively. Additional sugars are added in the Golgi compartment during passage through the secretory pathway to the plasma membrane. Of particular significance for Notch signaling is the addition of GlcNAc to O-fucose on an EGF repeat by the Fringe GlcNAc-transferases LFNG, MFNG or RFNG. Canonical Notch ligands (DLL1, DLL4, JAG1, JAG2) expressed in stromal cells bind to the extracellular domain of Notch receptors expressed in hematopoietic stem cells and myeloid and lymphoid progenitors to activate Notch signaling. Ligand-receptor binding is differentially regulated by the O-glycans on Notch. This review will summarize our understanding of the regulation of Notch signaling in myeloid and lymphoid cell development by specific O-glycans in mice with dysregulated expression of a particular glycosyltransferase and discuss how this may impact immune system development and malignancy in general, and in individuals with a congenital defect in the synthesis of the O-glycans attached to EGF repeats.
Collapse
Affiliation(s)
- Pamela Stanley
- Department of Cell Biology, Albert Einstein College Medicine, New York, NY, United States
| | | |
Collapse
|
5
|
Marimuthu S, Rauth S, Ganguly K, Zhang C, Lakshmanan I, Batra SK, Ponnusamy MP. Mucins reprogram stemness, metabolism and promote chemoresistance during cancer progression. Cancer Metastasis Rev 2021; 40:575-588. [PMID: 33813658 PMCID: PMC9635594 DOI: 10.1007/s10555-021-09959-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
Mucins are high-molecular-weight glycoproteins dysregulated in aggressive cancers. The role of mucins in disease progression, tumor proliferation, and chemotherapy resistance has been studied extensively. This article provides a comprehensive review of mucin's function as a physical barrier and the implication of mucin overexpression in impeded drug delivery to solid tumors. Mucins regulate the epithelial to mesenchymal transition (EMT) of cancer cells via several canonical and non-canonical oncogenic signaling pathways. Furthermore, mucins play an extensive role in enriching and maintaining the cancer stem cell (CSC) population, thereby sustaining the self-renewing and chemoresistant cellular pool in the bulk tumor. It has recently been demonstrated that mucins regulate the metabolic reprogramming during oncogenesis and cancer progression, which account for tumor cell survival, proliferation, and drug-resistance. This review article focuses on delineating mucin's role in oncogenic signaling and aberrant regulation of gene expressions, culminating in CSC maintenance, metabolic rewiring, and development of chemoresistance, tumor progression, and metastasis.
Collapse
Affiliation(s)
- Saravanakumar Marimuthu
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Chunmeng Zhang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Imayavaramban Lakshmanan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Wang L, Yu S, Chan ER, Chen KY, Liu C, Che D, Awadallah A, Myers J, Askew D, Huang AY, Maillard I, Huang D, Xin W, Zhou L. Notch-Regulated Dendritic Cells Restrain Inflammation-Associated Colorectal Carcinogenesis. Cancer Immunol Res 2021; 9:348-361. [PMID: 33441309 DOI: 10.1158/2326-6066.cir-20-0428] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/24/2020] [Accepted: 01/08/2021] [Indexed: 11/16/2022]
Abstract
Conventional dendritic cells (cDC) play a central role in T-cell antitumor responses. We studied the significance of Notch-regulated DC immune responses in a mouse model of colitis-associated colorectal cancer in which there is epithelial downregulation of Notch/Hes1 signaling. This defect phenocopies that caused by GMDS (GDP-mannose 4,6-dehydratase) mutation in human colorectal cancers. We found that, although wild-type immune cells restrained dysplasia progression and decreased the incidence of adenocarcinoma in chimeric mice, the immune system with Notch2 deleted in all blood lineages or in only DCs promoted inflammation-associated transformation. Notch2 signaling deficiency not only impaired cDC terminal differentiation, but also downregulated CCR7 expression, reduced DC migration, and suppressed antigen cross-presentation to CD8+ T cells. Transfer of Notch-primed DCs restrained inflammation-associated dysplasia progression. Consistent with the mouse data, we observed a correlation between infiltrating cDC1 and Notch2 signaling in human colorectal cancers and found that GMDS-mutant colorectal cancers showed decreased CCR7 expression and suppressed cDC1 signature gene expression. Suppressed cDC1 gene signature expression in human colorectal cancer was associated with a poor prognosis. In summary, our study supports an important role for Notch2 signaling in cDC1-mediated antitumor immunity and indicates that Notch2-controlled DCs restrain inflammation-associated colon cancer development in mice.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Shuiliang Yu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Ernest R Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | | | - Cui Liu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Danian Che
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Amad Awadallah
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Jay Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - David Askew
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Ivan Maillard
- Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio.,Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio. .,Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
7
|
Zhang S, Yang Q, Liu Z, Shi S. POFUT1 is dispensable for structure, function and survival of mouse podocytes. Am J Transl Res 2020; 12:2212-2224. [PMID: 32509213 PMCID: PMC7270003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 06/11/2023]
Abstract
Pofut1 gene encodes a O-fucosyltransferase that adds fucose to the serine/threonine residue in the sequence of C2XXXX(S/T)C3 of EGF-like domain in a protein. O-fucosylation has been shown to be required for some EGF-like domain-containing proteins to function, e.g., Notch1, and POFUT1 deficiency could affect cellular function and cause diseases. Pofut1 is ubiquitously expressed, but its essentiality for most cell types is not known. In the present study, we examined the consequence of Pofut1 gene abrogation in mouse podocytes using Cre-loxP system, and found that the conditional knockout mice were indistinguishable from wild-type controls in urinary protein level, glomerular morphology, podocyte foot process ultrastructure, podocyte marker expression and podocyte numbers. These results indicated that POFUT1 is not essential for podocyte structure, function and survival in mice. To understand why POFUT1 is dispensable for podocytes, we searched mouse podocyte essential gene candidates (as determined by single-cell RNA-seq) and found only two POFUT1 substrates, NOTCH2 and tPA. It has been shown that abrogation of these genes does not cause podocyte injury, explaining dispensability of POFUT1 for mouse podocytes and demonstrating a feasibility to predict POFUT1 essentiality for a given cell type. At present, most mouse cell types have been subject to single-cell RNA-seq, making essential gene prediction and thus POFUT1 requirement prediction possible for the cell types.
Collapse
Affiliation(s)
- Sipan Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of MedicineNanjing, China
| | - Qianqian Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of MedicineNanjing, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of MedicineNanjing, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of MedicineNanjing, China
- Department of Medicine, Icahn School of Medicine at Mount SinaiUSA
| |
Collapse
|
8
|
Barkeer S, Chugh S, Batra SK, Ponnusamy MP. Glycosylation of Cancer Stem Cells: Function in Stemness, Tumorigenesis, and Metastasis. Neoplasia 2018; 20:813-825. [PMID: 30015157 PMCID: PMC6037882 DOI: 10.1016/j.neo.2018.06.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 02/06/2023] Open
Abstract
Aberrant glycosylation plays a critical role in tumor aggressiveness, progression, and metastasis. Emerging evidence associates cancer initiation and metastasis to the enrichment of cancer stem cells (CSCs). Several universal markers have been identified for CSCs characterization; however, a specific marker has not yet been identified for different cancer types. Specific glycosylation variation plays a major role in the progression and metastasis of different cancers. Interestingly, many of the CSC markers are glycoproteins and undergo differential glycosylation. Given the importance of CSCs and altered glycosylation in tumorigenesis, the present review will discuss current knowledge of altered glycosylation of CSCs and its application in cancer research.
Collapse
Affiliation(s)
- Srikanth Barkeer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE.
| | - Seema Chugh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
9
|
Recognition of EGF-like domains by the Notch-modifying O-fucosyltransferase POFUT1. Nat Chem Biol 2017; 13:757-763. [DOI: 10.1038/nchembio.2381] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/21/2017] [Indexed: 01/17/2023]
|
10
|
Glycosyltransferase ST6GAL1 contributes to the regulation of pluripotency in human pluripotent stem cells. Sci Rep 2015; 5:13317. [PMID: 26304831 PMCID: PMC4548446 DOI: 10.1038/srep13317] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/21/2015] [Indexed: 01/12/2023] Open
Abstract
Many studies have suggested the significance of glycosyltransferase-mediated macromolecule glycosylation in the regulation of pluripotent states in human pluripotent stem cells (hPSCs). Here, we observed that the sialyltransferase ST6GAL1 was preferentially expressed in undifferentiated hPSCs compared to non-pluripotent cells. A lectin which preferentially recognizes α-2,6 sialylated galactosides showed strong binding reactivity with undifferentiated hPSCs and their glycoproteins, and did so to a much lesser extent with differentiated cells. In addition, downregulation of ST6GAL1 in undifferentiated hPSCs led to a decrease in POU5F1 (also known as OCT4) protein and significantly altered the expression of many genes that orchestrate cell morphogenesis during differentiation. The induction of cellular pluripotency in somatic cells was substantially impeded by the shRNA-mediated suppression of ST6GAL1, partially through interference with the expression of endogenous POU5F1 and SOX2. Targeting ST6GAL1 activity with a sialyltransferase inhibitor during cell reprogramming resulted in a dose-dependent reduction in the generation of human induced pluripotent stem cells (hiPSCs). Collectively, our data indicate that ST6GAL1 plays an important role in the regulation of pluripotency and differentiation in hPSCs, and the pluripotent state in human cells can be modulated using pharmacological tools to target sialyltransferase activity.
Collapse
|
11
|
Wang YC, Lin V, Loring JF, Peterson SE. The 'sweet' spot of cellular pluripotency: protein glycosylation in human pluripotent stem cells and its applications in regenerative medicine. Expert Opin Biol Ther 2015; 15:679-87. [PMID: 25736263 DOI: 10.1517/14712598.2015.1021329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Human pluripotent stem cells (hPSCs) promise for the future of regenerative medicine. The structural and biochemical diversity associated with glycans makes them a unique type of macromolecule modification that is involved in the regulation of a vast array of biochemical events and cellular activities including pluripotency in hPSCs. The primary focus of this review article is to highlight recent advances in stem cell research from a glycobiological perspective. We also discuss how our understanding of glycans and glycosylation may help overcome barriers hindering the clinical application of hPSC-derived cells. AREAS COVERED A literature survey using NCBI-PubMed and Google Scholar was performed in 2014. EXPERT OPINION Regenerative medicine hopes to provide novel strategies to combat human disease and tissue injury that currently lack effective therapies. Although progress in this field is accelerating, many critical issues remain to be addressed in order for cell-based therapy to become a practical and safe treatment option. Emerging evidence suggests that protein glycosylation may significantly influence the regulation of cellular pluripotency, and that the exploitation of protein glycosylation in hPSCs and their differentiated derivatives may lead to transformative and translational discoveries for regenerative medicine. In addition, hPSCs represent a novel research platform for investigating glycosylation-related disease.
Collapse
Affiliation(s)
- Yu-Chieh Wang
- The University of North Texas Health Science Center, Department of Pharmaceutical Sciences , 3500 Camp Bowie Boulevard, RES-314G, Fort Worth, TX 76107 , USA +1 817 735 2944 ; +1 817 735 2603 ;
| | | | | | | |
Collapse
|
12
|
Freeze HH, Chong JX, Bamshad MJ, Ng BG. Solving glycosylation disorders: fundamental approaches reveal complicated pathways. Am J Hum Genet 2014; 94:161-75. [PMID: 24507773 DOI: 10.1016/j.ajhg.2013.10.024] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Indexed: 11/30/2022] Open
Abstract
Over 100 human genetic disorders result from mutations in glycosylation-related genes. In 2013, a new glycosylation disorder was reported every 17 days. This trend will probably continue given that at least 2% of the human genome encodes glycan-biosynthesis and -recognition proteins. Established biosynthetic pathways provide many candidate genes, but finding unanticipated mutated genes will offer new insights into glycosylation. Simple glycobiomarkers can be used in narrowing the candidates identified by exome and genome sequencing, and those can be validated by glycosylation analysis of serum or cells from affected individuals. Model organisms will expand the understanding of these mutations' impact on glycosylation and pathology. Here, we highlight some recently discovered glycosylation disorders and the barriers, breakthroughs, and surprises they presented. We predict that some glycosylation disorders might occur with greater frequency than current estimates of their prevalence. Moreover, the prevalence of some disorders differs substantially between European and African Americans.
Collapse
Affiliation(s)
- Hudson H Freeze
- Human Genetics Program, Sanford Children's Health Research Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| | - Jessica X Chong
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Bobby G Ng
- Human Genetics Program, Sanford Children's Health Research Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
Hernandez Tejada FN, Galvez Silva JR, Zweidler-McKay PA. The challenge of targeting notch in hematologic malignancies. Front Pediatr 2014; 2:54. [PMID: 24959528 PMCID: PMC4051192 DOI: 10.3389/fped.2014.00054] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/21/2014] [Indexed: 01/12/2023] Open
Abstract
Notch signaling can play oncogenic and tumor suppressor roles depending on cell type. Hematologic malignancies encompass a wide range of transformed cells, and consequently the roles of Notch are diverse in these diseases. For example Notch is a potent T-cell oncogene, with >50% of T-cell acute lymphoblastic leukemia (T-ALL) cases carry activating mutations in the Notch1 receptor. Targeting Notch signaling in T-ALL with gamma-secretase inhibitors, which prevent Notch receptor activation, has shown pre-clinical activity, and is under evaluation clinically. In contrast, Notch signaling inhibits acute myeloblastic leukemia growth and survival, and although targeting Notch signaling in AML with Notch activators appears to have pre-clinical activity, no Notch agonists are clinically available at this time. As such, despite accumulating evidence about the biology of Notch signaling in different hematologic cancers, which provide compelling clinical promise, we are only beginning to target this pathway clinically, either on or off. In this review, we will summarize the evidence for oncogenic and tumor suppressor roles of Notch in a wide range of leukemias and lymphomas, and describe therapeutic opportunities for now and the future.
Collapse
Affiliation(s)
| | - Jorge R Galvez Silva
- Department of Pediatrics, University of Texas M. D. Anderson Cancer Center , Houston, TX , USA
| | | |
Collapse
|
14
|
Abstract
Post-translational modifications (PTMs) are known to be essential mechanisms used by eukaryotic cells to diversify their protein functions and dynamically coordinate their signaling networks. Defects in PTMs have been linked to numerous developmental disorders and human diseases, highlighting the importance of PTMs in maintaining normal cellular states. Human pluripotent stem cells (hPSCs), including embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), are capable of self-renewal and differentiation into a variety of functional somatic cells; these cells hold a great promise for the advancement of biomedical research and clinical therapy. The mechanisms underlying cellular pluripotency in human cells have been extensively explored in the past decade. In addition to the vast amount of knowledge obtained from the genetic and transcriptional research in hPSCs, there is a rapidly growing interest in the stem cell biology field to examine pluripotency at the protein and PTM level. This review addresses recent progress toward understanding the role of PTMs (glycosylation, phosphorylation, acetylation and methylation) in the regulation of cellular pluripotency.
Collapse
|
15
|
Myeloproliferation and hematopoietic stem cell dysfunction due to defective Notch receptor modification by O-fucose glycans. Semin Immunopathol 2012; 34:455-69. [DOI: 10.1007/s00281-012-0303-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 02/24/2012] [Indexed: 02/01/2023]
|
16
|
Beck RC. Production of cytotoxic, KIR-negative NK cells from CD34+ cord blood cells with the use of Notch signaling. Transfusion 2012; 51 Suppl 4:145S-152S. [PMID: 22074625 DOI: 10.1111/j.1537-2995.2011.03377.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The use of natural killer (NK) cells as cell therapy against acute leukemia is an active area of investigation. The optimal source of cytotoxic NK cells for therapeutic use is presently unknown. With funds from the National Blood Foundation, the author's lab has developed in vitro culture systems that use the Notch receptor ligand Delta4 for the differentiation and expansion of functional NK cells from CD34+ cord blood hematopoietic progenitor cells. These Notch-induced NK (N-NK) cells display a predominantly immature, CD56(bright) surface phenotype, with expression of activating receptors important for leukemia cell recognition and killing, but with an absence of inhibitory receptors that bind major histocompatibility complex (MHC) class I, making them free of restriction by self-MHC. They are capable of directly killing hematopoietic tumor cell lines and primary leukemia cells in vitro. Thus, cytotoxic, HLA-independent N-NK cells may represent a novel cell therapy for hematopoietic malignancy.
Collapse
Affiliation(s)
- Rose C Beck
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
17
|
Protein O-fucosyltransferase 1 (Pofut1) regulates lymphoid and myeloid homeostasis through modulation of Notch receptor ligand interactions. Blood 2011; 117:5652-62. [PMID: 21464368 DOI: 10.1182/blood-2010-12-326074] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Notch signaling is essential for lymphocyte development and is also implicated in myelopoiesis. Notch receptors are modified by O-fucosylation catalyzed by protein O-fucosyltransferase 1 (Pofut1). Fringe enzymes add N-acetylglucosamine to O-fucose and modify Notch signaling by altering the sensitivity of Notch receptors to Notch ligands. To address physiologic functions in hematopoiesis of Notch modified by O-fucose glycans, we examined mice with inducible inactivation of Pofut1 using Mx-Cre. These mice exhibited a reduction in T lymphopoiesis and in the production of marginal-zone B cells, in addition to myeloid hyperplasia. Restoration of Notch1 signaling rescued T lymphopoiesis and the marrow myeloid hyperplasia. After marrow transfer, both cell-autonomous and environmental cues were found to contribute to lymphoid developmental defects and myeloid hyperplasia in Pofut1-deleted mice. Although Pofut1 deficiency slightly decreased cell surface expression of Notch1 and Notch2, it completely abrogated the binding of Notch receptors with Delta-like Notch ligands and suppressed downstream Notch target activation, indicating that O-fucose glycans are critical for efficient Notch-ligand binding that transduce Notch signals. The combined data support a key role for the O-fucose glycans generated by Pofut1 in Notch regulation of hematopoietic homeostasis through modulation of Notch-ligand interactions.
Collapse
|
18
|
Myers J, Huang Y, Wei L, Yan Q, Huang A, Zhou L. Fucose-deficient hematopoietic stem cells have decreased self-renewal and aberrant marrow niche occupancy. Transfusion 2011; 50:2660-9. [PMID: 20573072 DOI: 10.1111/j.1537-2995.2010.02745.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Modification of Notch receptors by O-linked fucose and its further elongation by the Fringe family of glycosyltransferase has been shown to be important for Notch signaling activation. Our recent studies disclose a myeloproliferative phenotype, hematopoietic stem cell (HSC) dysfunction, and abnormal Notch signaling in mice deficient in FX, which is required for fucosylation of a number of proteins including Notch. The purpose of this study was to assess the self-renewal and stem cell niche features of fucose-deficient HSCs. STUDY DESIGN AND METHODS Homeostasis and maintenance of HSCs derived from FX(-/-) mice were studied by serial bone marrow transplantation, homing assay, and cell cycle analysis. Two-photon intravital microscopy was performed to visualize and compare the in vivo marrow niche occupancy by fucose-deficient and wild-type (WT) HSCs. RESULTS Marrow progenitors from FX(-/-) mice had mild homing defects that could be partially prevented by exogenous fucose supplementation. Fucose-deficient HSCs from FX(-/-) mice displayed decreased self-renewal capability compared with the WT controls. This is accompanied with their increased cell cycling activity and suppressed Notch ligand binding. When tracked in vivo by two-photon intravital imaging, the fucose-deficient HSCs were found localized farther from the endosteum of the calvarium marrow than the WT HSCs. CONCLUSIONS The current reported aberrant niche occupancy by HSCs from FX(-/-) mice, in the context of a faulty blood lineage homeostasis and HSC dysfunction in mice expressing Notch receptors deficient in O-fucosylation, suggests that fucosylation-modified Notch receptor may represent a novel extrinsic regulator for HSC engraftment and HSC niche maintenance.
Collapse
Affiliation(s)
- Jay Myers
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | |
Collapse
|