1
|
Lopes AL, Paulino AC, Thaumaturgo MAS, Araújo WM, Caloba P, Kawanishi K, Willert K, De Oliveira RP, Machado JC, Lemos F, Varki N, Morgado-Diaz JA, Läubli H, Todeschini AR, Varki A, Alisson-Silva F. Dietary intake of the red meat-derived glycan Neu5Gc fuels colorectal cancer through up-regulation of Wnt signaling pathway. Cancer Lett 2025; 616:217598. [PMID: 40023392 DOI: 10.1016/j.canlet.2025.217598] [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: 10/16/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Colorectal cancer (CRC) is a significant health concern, often linked to western diets, particularly red meat consumption. Several mechanisms, such as the high heme iron content, the formation of N-nitroso compounds (NOCs), heterocyclic amines (HCAs), and polycyclic aromatic hydrocarbons (PAHs), have been suggested to explain red meat's cancer-promoting effects. However, these factors are also found in fish and poultry, which are not linked to CRC risk. A new hypothesis attributes red meat's impact on CRC to its high content of a nonhuman glycan. While most mammals express N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), humans express only Neu5Ac due to the loss of the CMAH enzyme. A red meat-rich diet leads to the incorporation of Neu5Gc into human cells, triggering an antibody-mediated inflammatory process known as xenosialitis. This study shows that Neu5Gc incorporation into CRC cells activates the Wnt/β-catenin signaling pathway, promoting cell proliferation. In a murine model lacking CMAH, a Neu5Gc-enriched diet induced intestinal polyp growth, with more malignant characteristics. Additionally, Neu5Gc incorporation in intestinal cells increased the expression of genes downstream of Wnt signaling. These findings reveal, for the first time in an in vivo model, a mechanism independent of immune response, where red meat consumption accelerates tumor progression through Neu5Gc incorporation. This activation of the Wnt/β-catenin signaling pathway provides new insight into how red meat consumption may influence CRC progression.
Collapse
Affiliation(s)
- Ana Luiza Lopes
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Amanda Carlos Paulino
- Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Mariana A S Thaumaturgo
- Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | | | - Philippe Caloba
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil
| | - Kunio Kawanishi
- Department of Anatomy, Showa Medical University, Tokyo, Japan
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California San Diego, UCSD, USA
| | | | - João C Machado
- Biomedical Engineering Program, COPPE/UFRJ Brazil, Brazil; Post-Graduation Program on Surgical Sciences, School of Medicine/UFRJ, Brazil
| | - Felipe Lemos
- Immunopharmacology Laboratory, Fiocruz Brazil, Brazil
| | - Nissi Varki
- Departments of Medicine and Cellular & Molecular Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Glycobiology Research and Training Center (GRTC), University of California San Diego, UCSD, USA
| | | | - Heinz Läubli
- Division of Oncology and Department of Biomedicine, University Hospital and University of Basel, 4031, Basel, Switzerland
| | | | - Ajit Varki
- Departments of Medicine and Cellular & Molecular Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Glycobiology Research and Training Center (GRTC), University of California San Diego, UCSD, USA
| | - Frederico Alisson-Silva
- Department of Immunology, Paulo de Góes Institute of Microbiology, IMPG/UFRJ, Brazil; Structural and Functional Glycobiology Laboratory (LaGEF) - Carlos Chagas Filho Institute of Biophysics, IBCCF/UFRJ, Brazil.
| |
Collapse
|
2
|
Wang J, Shewell LK, Day CJ, Jennings MP. N-glycolylneuraminic acid as a carbohydrate cancer biomarker. Transl Oncol 2023; 31:101643. [PMID: 36805917 PMCID: PMC9971276 DOI: 10.1016/j.tranon.2023.101643] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/31/2023] [Accepted: 02/11/2023] [Indexed: 02/20/2023] Open
Abstract
One of the forms of aberrant glycosylation in human tumors is the expression of N-glycolylneuraminic acid (Neu5Gc). The only known enzyme to biosynthesize Neu5Gc in mammals, cytidine-5'-monophosphate-N-acetylneuraminic acid (CMAH), appears to be genetically inactivated in humans. Regardless, low levels of Neu5Gc have been detected in healthy humans. Therefore, it is proposed that the presence of Neu5Gc in humans is from dietary acquisition, such as red meat. Notably, detection of elevated Neu5Gc levels has been repeatedly found in cancer tissues, cells and serum samples, thereby Neu5Gc-containing antigens may be exploited as a class of cancer biomarkers. Here we review the findings to date on using Neu5Gc-containing tumor glycoconjugates as a class of cancer biomarkers for cancer detection, surveillance, prognosis and therapeutic targets. We review the evidence that supports an emerging hypothesis of de novo Neu5Gc biosynthesis in human cancer cells as a source of Neu5Gc in human tumors, generated under certain metabolic conditions.
Collapse
Affiliation(s)
- Jing Wang
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Lucy K Shewell
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | | | | |
Collapse
|
3
|
Gupta S, Shah B, Fung CS, Chan PK, Wakefield DL, Kuhns S, Goudar CT, Piret JM. Engineering protein glycosylation in CHO cells to be highly similar to murine host cells. Front Bioeng Biotechnol 2023; 11:1113994. [PMID: 36873370 PMCID: PMC9978007 DOI: 10.3389/fbioe.2023.1113994] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Since 2015 more than 34 biosimilars have been approved by the FDA. This new era of biosimilar competition has stimulated renewed technology development focused on therapeutic protein or biologic manufacturing. One challenge in biosimilar development is the genetic differences in the host cell lines used to manufacture the biologics. For example, many biologics approved between 1994 and 2011 were expressed in murine NS0 and SP2/0 cell lines. Chinese Hamster ovary (CHO) cells, however, have since become the preferred hosts for production due to their increased productivity, ease of use, and stability. Differences between murine and hamster glycosylation have been identified in biologics produced using murine and CHO cells. In the case of monoclonal antibodies (mAbs), glycan structure can significantly affect critical antibody effector function, binding activity, stability, efficacy, and in vivo half-life. In an attempt to leverage the intrinsic advantages of the CHO expression system and match the reference biologic murine glycosylation, we engineered a CHO cell expressing an antibody that was originally produced in a murine cell line to produce murine-like glycans. Specifically, we overexpressed cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) and N-acetyllactosaminide alpha-1,3-galactosyltransferase (GGTA) to obtain glycans with N-glycolylneuraminic acid (Neu5Gc) and galactose-α-1,3-galactose (alpha gal). The resulting CHO cells were shown to produce mAbs with murine glycans, and they were then analyzed by the spectrum of analytical methods typically used to demonstrate analytical similarity as a part of demonstrating biosimilarity. This included high-resolution mass spectrometry, biochemical, as well as cell-based assays. Through selection and optimization in fed-batch cultures, two CHO cell clones were identified with similar growth and productivity criteria to the original cell line. They maintained stable production for 65 population doubling times while matching the glycosylation profile and function of the reference product expressed in murine cells. This study demonstrates the feasibility of engineering CHO cells to express mAbs with murine glycans to facilitate the development of biosimilars that are highly similar to marketed reference products expressed in murine cells. Furthermore, this technology can potentially reduce the residual uncertainty regarding biosimilarity, resulting in a higher probability of regulatory approval and potentially reduced costs and time in development.
Collapse
Affiliation(s)
- Shivani Gupta
- Amgen, Inc., San Francisco, CA, United States.,Michael Smith Laboratories, and Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | - Scott Kuhns
- Amgen, Inc., Thousand Oaks, CA, United States
| | | | - James M Piret
- Michael Smith Laboratories, and Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
CMAHP promotes metastasis by reducing ubiquitination of Snail and inducing angiogenesis via GM-CSF overexpression in gastric cancer. Oncogene 2022; 41:159-172. [PMID: 34716430 DOI: 10.1038/s41388-021-02087-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/18/2022]
Abstract
Pseudogenes are generally considered "junk" DNA or "genomic fossils" generated during the evolution process that lack biological activity. However, accumulating reports indicate that pseudogenes have biological functions critical for cancer development. Experiments from the current study showed marked overexpression of the cytidine monophospho-N-acetylneuraminic acid hydroxylase pseudogene (CMAHP) in gastric cancer, which was associated with poor overall survival. However, the mechanisms underlying the activity of CMAHP in tumor development are largely unknown. Gene Set Enrichment Analysis (GSEA) revealed that CMAHP-correlated genes are significantly involved in epithelial-mesenchymal transition (EMT) and angiogenesis. Functional studies further confirmed that CMAHP mediates metastasis and angiogenesis in vitro and in vivo. Furthermore, CMAHP promoted cancer cell migration, invasion, and metastasis through Snail overexpression, which decreased ubiquitination mediated by NF-κB signaling. Angiogenesis is known to be induced by granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulation. CMAHP increased GM-CSF transactivation via promoting direct binding of c-Jun to the -1981/-1975 region of the GM-CSF promoter. Notably, CMAHP interacts with Histone H1.4 promoting histone acetylation to enhance c-Jun and RelA (p65) expression. Our collective findings provide novel evidence that CMAHP contributes to tumor progression and modulates metastasis and angiogenesis in gastric cancer.
Collapse
|
5
|
Wood CL, Suchacki KJ, van 't Hof R, Cawthorn WP, Dillon S, Straub V, Wong SC, Ahmed SF, Farquharson C. A comparison of the bone and growth phenotype of mdx, mdx:Cmah-/- and mdx:Utrn +/- murine models with the C57BL/10 wild-type mouse. Dis Model Mech 2020; 13:dmm.040659. [PMID: 31754018 PMCID: PMC6994935 DOI: 10.1242/dmm.040659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
The muscular dystrophy X-linked (mdx) mouse is commonly used as a mouse model of Duchenne muscular dystrophy (DMD). Its phenotype is, however, mild, and other mouse models have been explored. The mdx:Cmah−/− mouse carries a human-like mutation in the Cmah gene and has a severe muscle phenotype, but its growth and bone development are unknown. In this study, we compared male mdx, mdx:Utrn+/−, mdx:Cmah−/− and wild-type (WT) mice at 3, 5 and 7 weeks of age to determine the suitability of the mdx:Cmah−/− mouse as a model for assessing growth and skeletal development in DMD. The mdx:Cmah−/− mice were lighter than WT mice at 3 weeks, but heavier at 7 weeks, and showed an increased growth rate at 5 weeks. Cortical bone fraction as assessed by micro-computed tomography was greater in both mdx and mdx:Cmah−/− mice versus WT mice at 7 weeks. Tissue mineral density was also higher in mdx:Cmah−/− mice at 3 and 7 weeks. Gene profiling of mdx:Cmah−/− bone identified increased expression of Igf1, Igf1r and Vegfa. Both the mdx and mdx:Cmah−/− mice showed an increased proportion of regulated bone marrow adipose tissue (BMAT) but a reduction in constitutive BMAT. The mdx:Cmah−/− mice show evidence of catch-up growth and more rapid bone development. This pattern does not mimic the typical DMD growth trajectory and therefore the utility of the mdx:Cmah−/− mouse for studying growth and skeletal development in DMD is limited. Further studies of this model may, however, shed light on the phenomenon of catch-up growth. This article has an associated First Person interview with the first author of the paper. Summary: Unlike boys with DMD, the mdx:Cmah−/− mouse shows increased weight gain and more rapid bone development; therefore, its utility for studying growth and skeletal development in DMD is limited.
Collapse
Affiliation(s)
- Claire L Wood
- Division of Developmental Biology, Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, UK .,John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, NE1 3BZ, UK
| | - Karla J Suchacki
- BHF Centre for Cardiovascular Science, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Rob van 't Hof
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Will P Cawthorn
- BHF Centre for Cardiovascular Science, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Scott Dillon
- Division of Developmental Biology, Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, NE1 3BZ, UK
| | - Sze Choong Wong
- Developmental Endocrinology Research Group, School of Medicine, University of Glasgow, Glasgow, G51 4TF, UK
| | - Syed F Ahmed
- Developmental Endocrinology Research Group, School of Medicine, University of Glasgow, Glasgow, G51 4TF, UK
| | - Colin Farquharson
- Division of Developmental Biology, Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, UK
| |
Collapse
|
6
|
Salonius E, Kontturi L, Laitinen A, Haaparanta AM, Korhonen M, Nystedt J, Kiviranta I, Muhonen V. Chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells in a three-dimensional environment. J Cell Physiol 2019; 235:3497-3507. [PMID: 31552691 DOI: 10.1002/jcp.29238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Cell therapy combined with biomaterial scaffolds is used to treat cartilage defects. We hypothesized that chondrogenic differentiation bone marrow-derived mesenchymal stem cells (BM-MSCs) in three-dimensional biomaterial scaffolds would initiate cartilaginous matrix deposition and prepare the construct for cartilage regeneration in situ. The chondrogenic capability of human BM-MSCs was first verified in a pellet culture. The BM-MSCs were then either seeded onto a composite scaffold rhCo-PLA combining polylactide and collagen type II (C2) or type III (C3), or commercial collagen type I/III membrane (CG). The BM-MSCs were either cultured in a proliferation medium or chondrogenic culture medium. Adult human chondrocytes (ACs) served as controls. After 3, 14, and 28 days, the constructs were analyzed with quantitative polymerase chain reaction and confocal microscopy and sulfated glycosaminoglycans (GAGs) were measured. The differentiated BM-MSCs entered a hypertrophic state by Day 14 of culture. The ACs showed dedifferentiation with no expression of chondrogenic genes and low amount of GAG. The CG membrane induced the highest expression levels of hypertrophic genes. The two different collagen types in composite scaffolds yielded similar results. Regardless of the biomaterial scaffold, culturing BM-MSCs in chondrogenic differentiation medium resulted in chondrocyte hypertrophy. Thus, caution for cell fate is required when designing cell-biomaterial constructs for cartilage regeneration.
Collapse
Affiliation(s)
- Eve Salonius
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland
| | - Leena Kontturi
- Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Anne-Marie Haaparanta
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Johanna Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Ilkka Kiviranta
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland.,Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Virpi Muhonen
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Dagur RS, Branch-Woods A, Mathews S, Joshi PS, Quadros RM, Harms DW, Cheng Y, Miles SM, Pirruccello SJ, Gurumurthy CB, Gorantla S, Poluektova LY. Human-like NSG mouse glycoproteins sialylation pattern changes the phenotype of human lymphocytes and sensitivity to HIV-1 infection. BMC Immunol 2019; 20:2. [PMID: 30616506 PMCID: PMC6322283 DOI: 10.1186/s12865-018-0279-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/30/2018] [Indexed: 01/14/2023] Open
Abstract
Background The use of immunodeficient mice transplanted with human hematopoietic stem cells is an accepted approach to study human-specific infectious diseases such as HIV-1 and to investigate multiple aspects of human immune system development. However, mouse and human are different in sialylation patterns of proteins due to evolutionary mutations of the CMP-N-acetylneuraminic acid hydroxylase (CMAH) gene that prevent formation of N-glycolylneuraminic acid from N-acetylneuraminic acid. How changes in the mouse glycoproteins’ chemistry affect phenotype and function of transplanted human hematopoietic stem cells and mature human immune cells in the course of HIV-1 infection are not known. Results We mutated mouse CMAH in the NOD/scid-IL2Rγc−/− (NSG) mouse strain, which is widely used for the transplantation of human cells, using the CRISPR/Cas9 system. The new strain provides a better environment for human immune cells. Transplantation of human hematopoietic stem cells leads to broad B cells repertoire, higher sensitivity to HIV-1 infection, and enhanced proliferation of transplanted peripheral blood lymphocytes. The mice showed no effect on the clearance of human immunoglobulins and enhanced transduction efficiency of recombinant adeno-associated viral vector rAAV2/DJ8. Conclusion NSG-cmah−/− mice expand the mouse models suitable for human cells transplantation, and this new model has advantages in generating a human B cell repertoire. This strain is suitable to study different aspects of the human immune system development, provide advantages in patient-derived tissue and cell transplantation, and could allow studies of viral vectors and infectious agents that are sensitive to human-like sialylation of mouse glycoproteins. Electronic supplementary material The online version of this article (10.1186/s12865-018-0279-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raghubendra Singh Dagur
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Amanda Branch-Woods
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Saumi Mathews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Poonam S Joshi
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, Omaha, NE, USA
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, Omaha, NE, USA
| | - Donald W Harms
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, Omaha, NE, USA
| | - Yan Cheng
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | | | | | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research Office, Omaha, NE, USA.,Developmental Neuroscience, Munroe Meyer Institute for Genetics and Rehabilitation, of University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
8
|
Abstract
Tumor-associated gangliosides play important roles in regulation of signal transduction induced by growth-factor receptors including EGFR, FGFR, HGF and PDGFR in a specific microdomain called glycosynapse in the cancer cell membranes, and in interaction with glycan recognition molecules involved in cell adhesion and immune regulation including selectins and siglecs. As the genes involved in the synthesis and degradation of tumor-associated gangliosides were identified, biological functions became clearer from the experimental results employing forced overexpression and/or knockdown/knockout of the genes. Studies on the regulatory mechanisms for their expression also achieved great advancements. Epigenetic silencing of glycan-related genes is a dominant mechanism in glycan alteration at early stages of carcinogenesis. Development of hypoxia resistance involving activation of a transcription factor HIF, and acquisition of cancer stem cell-like characteristics through epithelial-mesenchymal transition are important mechanisms for glycan modulations in the later stages of cancer progression. In the initial stages of studies, the gangliosides which specifically appear in cancers attracted attention under the name of tumor-associated gangliosides. However, it became apparent that not only the cancer-associated gangliosides but also the normal gangliosides present in nonmalignant cells and tissues perform important biological functions, and some of them tend to disappear in cancer cells resulting in the loss of the physiological functions, and this sometimes facilitates progression of cancers.
Collapse
|
9
|
Okerblom J, Varki A. Biochemical, Cellular, Physiological, and Pathological Consequences of Human Loss of N-Glycolylneuraminic Acid. Chembiochem 2017; 18:1155-1171. [PMID: 28423240 DOI: 10.1002/cbic.201700077] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Indexed: 12/15/2022]
Abstract
About 2-3 million years ago, Alu-mediated deletion of a critical exon in the CMAH gene became fixed in the hominin lineage ancestral to humans, possibly through a stepwise process of selection by pathogen targeting of the CMAH product (the sialic acid Neu5Gc), followed by reproductive isolation through female anti-Neu5Gc antibodies. Loss of CMAH has occurred independently in some other lineages, but is functionally intact in Old World primates, including our closest relatives, the chimpanzee. Although the biophysical and biochemical ramifications of losing tens of millions of Neu5Gc hydroxy groups at most cell surfaces remains poorly understood, we do know that there are multiscale effects functionally relevant to both sides of the host-pathogen interface. Hominin CMAH loss might also contribute to understanding human evolution, at the time when our ancestors were starting to use stone tools, increasing their consumption of meat, and possibly hunting. Comparisons with chimpanzees within ethical and practical limitations have revealed some consequences of human CMAH loss, but more has been learned by using a mouse model with a human-like Cmah inactivation. For example, such mice can develop antibodies against Neu5Gc that could affect inflammatory processes like cancer progression in the face of Neu5Gc metabolic incorporation from red meats, display a hyper-reactive immune system, a human-like tendency for delayed wound healing, late-onset hearing loss, insulin resistance, susceptibility to muscular dystrophy pathologies, and increased sensitivity to multiple human-adapted pathogens involving sialic acids. Further studies in such mice could provide a model for other human-specific processes and pathologies involving sialic acid biology that have yet to be explored.
Collapse
Affiliation(s)
- Jonathan Okerblom
- Biomedical Sciences Graduate Program, University of California in San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0687, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, GRTC) and, Center for Academic Research and Training in Anthropogeny, CARTA), Departments of Medicine and Cellular and Molecular Medicine, University of California in San Diego, La Jolla, CA, 92093-0687, USA
| |
Collapse
|
10
|
Babbucci M, Ferraresso S, Pauletto M, Franch R, Papetti C, Patarnello T, Carnier P, Bargelloni L. An integrated genomic approach for the study of mandibular prognathism in the European seabass (Dicentrarchus labrax). Sci Rep 2016; 6:38673. [PMID: 27929136 PMCID: PMC5144136 DOI: 10.1038/srep38673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022] Open
Abstract
Skeletal anomalies in farmed fish are a relevant issue affecting animal welfare and health and causing significant economic losses. Here, a high-density genetic map of European seabass for QTL mapping of jaw deformity was constructed and a genome-wide association study (GWAS) was carried out on a total of 298 juveniles, 148 of which belonged to four full-sib families. Out of 298 fish, 107 were affected by mandibular prognathism (MP). Three significant QTLs and two candidate SNPs associated with MP were identified. The two GWAS candidate markers were located on ChrX and Chr17, both in close proximity with the peaks of the two most significant QTLs. Notably, the SNP marker on Chr17 was positioned within the Sobp gene coding region, which plays a pivotal role in craniofacial development. The analysis of differentially expressed genes in jaw-deformed animals highlighted the “nervous system development” as a crucial pathway in MP. In particular, Zic2, a key gene for craniofacial morphogenesis in model species, was significantly down-regulated in MP-affected animals. Gene expression data revealed also a significant down-regulation of Sobp in deformed larvae. Our analyses, integrating transcriptomic and GWA methods, provide evidence for putative mechanisms underlying seabass jaw deformity.
Collapse
Affiliation(s)
- Massimiliano Babbucci
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Serena Ferraresso
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Rafaella Franch
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Chiara Papetti
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35121 Padova, Italy
| | - Tomaso Patarnello
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Paolo Carnier
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| | - Luca Bargelloni
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro, Italy
| |
Collapse
|
11
|
Song KH, Kwak CH, Jin UH, Ha SH, Park JY, Abekura F, Chang YC, Cho SH, Lee K, Chung TW, Ha KT, Lee YC, Kim CH. Housekeeping promoter 5'pcmah-2 of pig CMP-N-acetylneuraminic acid hydroxylase gene for NeuGc expression. Glycoconj J 2016; 33:779-788. [PMID: 27188588 DOI: 10.1007/s10719-016-9671-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/12/2016] [Accepted: 05/02/2016] [Indexed: 01/21/2023]
Abstract
In the present study, we isolated pCMAH house-keeping promoter regions (Ph), which are responsible for transcriptional regulation and which are located upstream of the alternative transcript pcmah-2. Luciferase reporter assays using serial construction of each deleted promoter demonstrated that the Ph promoter was highly active in pig-derived kidney PK15. Ph promoter of pcmah lacked a TATA box, but contained three putative Sp1 binding sites. Mutations of these Sp1 binding sites always resulted in the reduction of luciferase activities in Ph-334. In addition, treatment with mithramycin A (25-100 nM) decreased the luciferase activities of the Ph promoters and NeuGc expression in a dose-dependent manner. Electrophoretic mobility shift assay analysis revealed that the probes containing each Sp1 binding site bound to Sp1. Taken together, the results indicate that Sp1 bind to their putative binding sites on the Ph promoter regions of pcmah and positively regulate the promoter activity in pig kidney cells. Interspecies comparison of 5'UTRs and 5'flanking regions shows high homology between pig and cattle, and Sp1 binding sites existing in genomic regions corresponding Ph region are evolutionally conserved.
Collapse
Affiliation(s)
- Kwon-Ho Song
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Choong-Hwan Kwak
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Un-Ho Jin
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Sun-Hyung Ha
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Jun-Young Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Fukushi Abekura
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, 705-034, South Korea
| | - Seung-Hak Cho
- Department of Enteric Bacterial Infections, Korea Centers for Disease Control and Prevention, Cheongwon-gun, South Korea
| | - Kichoon Lee
- Functional Genomics Laboratory, Department of Animal Sciences, The Ohio State University, 2029 Fyffe Court, Columbus, OH, 43210, USA
| | - Tae-Wook Chung
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, South Korea
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, South Korea
| | - Young-Choon Lee
- Faculty of Medicinal Biotechnology, Dong-A University, Busan, 604-714, South Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Science, Sungkyunkwan University, Chunchun-Dong, Suwon, 16419, South Korea.
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, South Korea.
- Department of Biological Science, Sungkyunkwan University, 2066, Seobu-Ro, Suwon, Gyunggi-Do, 16419, South Korea.
| |
Collapse
|
12
|
Skog MS, Nystedt J, Korhonen M, Anderson H, Lehti TA, Pajunen MI, Finne J. Expression of neural cell adhesion molecule and polysialic acid in human bone marrow-derived mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:113. [PMID: 27528376 PMCID: PMC4986182 DOI: 10.1186/s13287-016-0373-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/28/2016] [Accepted: 07/21/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND In order to develop novel clinical applications and to gain insights into possible therapeutic mechanisms, detailed molecular characterization of human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) is needed. Neural cell adhesion molecule (NCAM, CD56) is a transmembrane glycoprotein modulating cell-cell and cell-matrix interactions. An additional post-translational modification of NCAM is the α2,8-linked polysialic acid (polySia). Because of its background, NCAM is often considered a marker of neural lineage commitment. Generally, hBM-MSCs are considered to be devoid of NCAM expression, but more rigorous characterization is needed. METHODS We have studied NCAM and polySia expression in five hBM-MSC lines at mRNA and protein levels. Cell surface localization was confirmed by immunofluorescence staining and expression frequency in the donor-specific lines by flow cytometry. For the detection of poorly immunogenic polySia, a fluorochrome-tagged catalytically defective enzyme was employed. RESULTS All five known NCAM isoforms are expressed in these cells at mRNA level and the three main isoforms are present at protein level. Both polysialyltransferases, generally responsible for NCAM polysialylation, are expressed at mRNA level, but only very few cells express polySia at the cell surface. CONCLUSIONS Our results underline the need for a careful control of methods and conditions in the characterization of MSCs. This study shows that, against the generally held view, clinical-grade hBM-MSCs do express NCAM. In contrast, although both polysialyltransferase genes are transcribed in these cells, very few express polySia at the cell surface. NCAM and polySia represent new candidate molecules for influencing MSC interactions.
Collapse
Affiliation(s)
- Maria S Skog
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| | - Johanna Nystedt
- Cell Therapy Services, Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310, Helsinki, Finland
| | - Matti Korhonen
- Cell Therapy Services, Finnish Red Cross Blood Service, Kivihaantie 7, FI-00310, Helsinki, Finland
| | - Heidi Anderson
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.,Present Address: Genoscoper Laboratories Oy, P.O. Box 1040, FI-00251, Helsinki, Finland
| | - Timo A Lehti
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Maria I Pajunen
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.,Present Address: Department of Bacteriology and Immunology, Medicum, Research Programs Unit, Immunobiology, University of Helsinki, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Jukka Finne
- Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| |
Collapse
|
13
|
Cusanovich DA, Caliskan M, Billstrand C, Michelini K, Chavarria C, De Leon S, Mitrano A, Lewellyn N, Elias JA, Chupp GL, Lang RM, Shah SJ, Decara JM, Gilad Y, Ober C. Integrated analyses of gene expression and genetic association studies in a founder population. Hum Mol Genet 2016; 25:2104-2112. [PMID: 26931462 PMCID: PMC5062579 DOI: 10.1093/hmg/ddw061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 02/21/2016] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWASs) have become a standard tool for dissecting genetic contributions to disease risk. However, these studies typically require extraordinarily large sample sizes to be adequately powered. Strategies that incorporate functional information alongside genetic associations have proved successful in increasing GWAS power. Following this paradigm, we present the results of 20 different genetic association studies for quantitative traits related to complex diseases, conducted in the Hutterites of South Dakota. To boost the power of these association studies, we collected RNA-sequencing data from lymphoblastoid cell lines for 431 Hutterite individuals. We then used Sherlock, a tool that integrates GWAS and expression quantitative trait locus (eQTL) data, to identify weak GWAS signals that are also supported by eQTL data. Using this approach, we found novel associations with quantitative phenotypes related to cardiovascular disease, including carotid intima-media thickness, left atrial volume index, monocyte count and serum YKL-40 levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jack A Elias
- Division of Biology and Medicine, Brown University, Providence, RI 02912, USA and
| | - Geoffrey L Chupp
- Pulmonary and Critical Care, Yale School of Medicine, New Haven, CT 06519, USA
| | - Roberto M Lang
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | - Sanjiv J Shah
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | - Jeanne M Decara
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
14
|
The Chemical Biology of Human Metallo-β-Lactamase Fold Proteins. Trends Biochem Sci 2016; 41:338-355. [PMID: 26805042 PMCID: PMC4819959 DOI: 10.1016/j.tibs.2015.12.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/19/2015] [Accepted: 12/22/2015] [Indexed: 01/30/2023]
Abstract
The αββα metallo β-lactamase (MBL) fold (MBLf) was first observed in bacterial enzymes that catalyze the hydrolysis of almost all β-lactam antibiotics, but is now known to be widely distributed. The MBL core protein fold is present in human enzymes with diverse biological roles, including cell detoxification pathways and enabling resistance to clinically important anticancer medicines. Human (h)MBLf enzymes can bind metals, including zinc and iron ions, and catalyze a range of chemically interesting reactions, including both redox (e.g., ETHE1) and hydrolytic processes (e.g., Glyoxalase II, SNM1 nucleases, and CPSF73). With a view to promoting basic research on MBLf enzymes and their medicinal targeting, here we summarize current knowledge of the mechanisms and roles of these important molecules. MBLs are mono- or di-zinc ion-dependent hydrolases that enable bacterial resistance to almost all β-lactam antibiotics. The αββα MBL core fold is widely distributed and supports a range of catalytic activities, including redox reactions. hMBL proteins are a small family of approximately 18 zinc- and iron-dependent proteins with roles in metabolism and/or detoxification and nucleic acid modification. In a notable parallel with the role of bacterial MBLs in antibiotic resistance, some hMBLf enzymes enable resistance to chemotherapy drugs, such as cisplatin and mitomycin C.
Collapse
|
15
|
Abstract
The sialic acids N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) differ by a single oxygen atom and are widely found at the terminal position of glycans on vertebrate cell surfaces. In animals capable of synthesizing Neu5Gc, most tissues and cell types express both sialic acids, in proportions that vary between species. However, it has long been noted that Neu5Gc is consistently expressed at trace to absent levels in the brains of all vertebrates studied to date. Although several reports have claimed to find low levels of Neu5Gc-containing glycans in neural tissue, no study definitively excludes the possibility of contamination with glycans from non-neural cell types. This distribution of a molecule - prominently but variably expressed in extraneural tissues but very low or absent in the brain - is, to our knowledge, unique. The evolutionarily conserved brain-specific suppression of Neu5Gc may indicate that its presence is toxic to this organ; however, no studies to date have directly addressed this very interesting question. Here we provide a historical background to this issue and discuss potential mechanisms causing the suppression of Neu5Gc expression in brain tissue, as well as mechanisms by which Neu5Gc may exert the presumed toxicity. Finally, we discuss future approaches towards understanding the mechanisms and implications of this unusual finding.
Collapse
Affiliation(s)
- Leela R L Davies
- Glycobiology Research and Training Center, Center for Academic Research and Training in Anthropogeny, Biomedical Sciences Graduate Program, Departments of Medicine and Cellular and Molecular Medicine, University of California at San Diego, 9500 Gilman Dr., MC 0687, La Jolla, CA, 92093-0687, USA
| | | |
Collapse
|
16
|
Cuccui J, Wren B. Hijacking bacterial glycosylation for the production of glycoconjugates, from vaccines to humanised glycoproteins. ACTA ACUST UNITED AC 2014; 67:338-50. [PMID: 25244672 DOI: 10.1111/jphp.12321] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/10/2014] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Glycosylation or the modification of a cellular component with a carbohydrate moiety has been demonstrated in all three domains of life as a basic post-translational process important in a range of biological processes. This review will focus on the latest studies attempting to exploit bacterial N-linked protein glycosylation for glycobiotechnological applications including glycoconjugate vaccine and humanised glycoprotein production. The challenges that remain for these approaches to reach full biotechnological maturity will be discussed. KEY FINDINGS Oligosaccharyltransferase-dependent N-linked glycosylation can be exploited to make glycoconjugate vaccines against bacterial pathogens. Few technical limitations remain, but it is likely that the technologies developed will soon be considered a cost-effective and flexible alternative to current chemical-based methods of vaccine production. Some highlights from current glycoconjugate vaccines developed using this in-vivo production system include a vaccine against Shigella dysenteriae O1 that has passed phase 1 clinical trials, a vaccine against the tier 1 pathogen Francisella tularensis that has shown efficacy in mice and a vaccine against Staphylococcus aureus serotypes 5 and 8. Generation of humanised glycoproteins within bacteria was considered impossible due to the distinct nature of glycan modification in eukaryotes and prokaryotes. We describe the method used to overcome this conundrum to allow engineering of a eukaryotic pentasaccharide core sugar modification within Escherichia coli. This core was assembled by combining the function of the initiating transferase WecA, several Alg genes from Saccharomyces cerevisiae and the oligosaccharyltransferase function of the Campylobacter jejuni PglB. Further exploitation of a cytoplasmic N-linked glycosylation system found in Actinobacillus pleuropneumoniae where the central enzyme is known as N-linking glycosyltransferase has overcome some of the limitations demonstrated by the oligosaccharyltransferase-dependent system. SUMMARY Characterisation of the first bacterial N-linked glycosylation system in the human enteropathogen Campylobacter jejuni has led to substantial biotechnological applications. Alternative methods for glycoconjugate vaccine production have been developed using this N-linked system. Vaccines against both Gram-negative and Gram-positive organisms have been developed, and efficacy testing has thus far demonstrated that the vaccines are safe and that robust immune responses are being detected. These are likely to complement and reduce the cost of current technologies thus opening new avenues for glycoconjugate vaccines. These new markets could potentially include glycoconjugate vaccines tailored specifically for animal vaccination, which has until today thought to be non-viable due to the cost of current in-vitro chemical conjugation methods. Utilisation of N-linked glycosylation to generate humanised glycoproteins is also close to becoming reality. This 'bottom up' assembly mechanism removes the heterogeneity seen in current humanised products. The majority of developments reported in this review exploit a single N-linked glycosylation system from Campylobacter jejuni; however, alternative N-linked glycosylation systems have been discovered which should help to overcome current technical limitations and perhaps more systems remain to be discovered. The likelihood is that further glycosylation systems exist and are waiting to be exploited.
Collapse
Affiliation(s)
- Jon Cuccui
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, UK
| | | |
Collapse
|
17
|
Tringali C, Silvestri I, Testa F, Baldassari P, Anastasia L, Mortarini R, Anichini A, López-Requena A, Tettamanti G, Venerando B. Molecular subtyping of metastatic melanoma based on cell ganglioside metabolism profiles. BMC Cancer 2014; 14:560. [PMID: 25085576 PMCID: PMC4132924 DOI: 10.1186/1471-2407-14-560] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/28/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In addition to alterations concerning the expression of oncogenes and onco-suppressors, melanoma is characterized by the presence of distinctive gangliosides (sialic acid carrying glycosphingolipids). Gangliosides strongly control cell surface dynamics and signaling; therefore, it could be assumed that these alterations are linked to modifications of cell behavior acquired by the tumor. On these bases, this work investigated the correlations between melanoma cell ganglioside metabolism profiles and the biological features of the tumor and the survival of patients. METHODS Melanoma cell lines were established from surgical specimens of AJCC stage III and IV melanoma patients. Sphingolipid analysis was carried out on melanoma cell lines and melanocytes through cell metabolic labeling employing [3-3H]sphingosine and by FACS. N-glycolyl GM3 was identified employing the 14 F7 antibody. Gene expression was assayed by Real Time PCR. Cell invasiveness was assayed through a Matrigel invasion assay; cell proliferation was determined through the soft agar assay, MTT, and [3H] thymidine incorporation. Statistical analysis was performed using XLSTAT software for melanoma hierarchical clustering based on ganglioside profile, the Kaplan-Meier method, the log-rank (Mantel-Cox) test, and the Mantel-Haenszel test for survival analysis. RESULTS Based on the ganglioside profiles, through a hierarchical clustering, we classified melanoma cells isolated from patients into three clusters: 1) cluster 1, characterized by high content of GM3, mainly in the form of N-glycolyl GM3, and GD3; 2) cluster 2, characterized by the appearance of complex gangliosides and by a low content of GM3; 3) cluster 3, which showed an intermediate phenotype between cluster 1 and cluster 3. Moreover, our data demonstrated that: a) a correlation could be traced between patients' survival and clusters based on ganglioside profiles, with cluster 1 showing the worst survival; b) the expression of several enzymes (sialidase NEU3, GM2 and GM1 synthases) involved in ganglioside metabolism was associated with patients' survival; c) melanoma clusters showed different malignant features such as growth in soft agar, invasiveness, expression of anti-apoptotic proteins. CONCLUSIONS Ganglioside profile and metabolism is strictly interconnected with melanoma aggressiveness. Therefore, the profiling of melanoma gangliosides and enzymes involved in their metabolism could represent a useful prognostic and diagnostic tool.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Bruno Venerando
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Milan, Italy.
| |
Collapse
|
18
|
Grassot V, Da Silva A, Saliba J, Maftah A, Dupuy F, Petit JM. Highlights of glycosylation and adhesion related genes involved in myogenesis. BMC Genomics 2014; 15:621. [PMID: 25051993 PMCID: PMC4223822 DOI: 10.1186/1471-2164-15-621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Myogenesis is initiated by myoblast differentiation and fusion to form myotubes and muscle fibres. A population of myoblasts, known as satellite cells, is responsible for post-natal growth of muscle and for its regeneration. This differentiation requires many changes in cell behaviour and its surrounding environment. These modifications are tightly regulated over time and can be characterized through the study of changes in gene expression associated with this process. During the initial myogenesis steps, using the myoblast cell line C2C12 as a model, Janot et al. (2009) showed significant variations in expression for genes involved in pathways of glycolipid synthesis. In this study we used murine satellite cells (MSC) and their ability to differentiate into myotubes or early fat storage cells to select glycosylation related genes whose variation of expression is myogenesis specific. RESULTS The comparison of variant genes in both MSC differentiation pathways identified 67 genes associated with myogenesis. Comparison with data obtained for C2C12 revealed that only 14 genes had similar expression profiles in both cell types and that 17 genes were specifically regulated in MSC. Results were validated statistically by without a priori clustering. Classification according to protein function encoded by these 31 genes showed that the main regulated cellular processes during this differentiation were (i) remodeling of the extracellular matrix, particularly, sulfated structures, (ii) down-regulation of O-mannosyl glycan biosynthesis, and (iii) an increase in adhesion protein expression. A functional study was performed on Itga11 and Chst5 encoding two highly up-regulated proteins. The inactivation of Chst5 by specific shRNA delayed the fusion of MSC. By contrast, the inactivation of Itga11 by specific shRNA dramatically decreased the fusion ability of MSC. This result was confirmed by neutralization of Itga11 product by specific antibodies. CONCLUSIONS Our screening method detected 31 genes specific for myogenic differentiation out of the 383 genes studied. According to their function, interaction networks of the products of these selected genes converged to cell fusion. Functional studies on Itga11 and Chst5 demonstrated the robustness of this screening.
Collapse
Affiliation(s)
- Vincent Grassot
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Anne Da Silva
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - James Saliba
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Abderrahman Maftah
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Fabrice Dupuy
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| | - Jean-Michel Petit
- INRA, UMR 1061 Unité de Génétique Moléculaire Animale, Université de Limoges, Faculté des Sciences et Techniques, 123 Avenue A. Thomas, Limoges 87060, France
| |
Collapse
|
19
|
Barone A, Säljö K, Benktander J, Blomqvist M, Månsson JE, Johansson BR, Mölne J, Aspegren A, Björquist P, Breimer ME, Teneberg S. Sialyl-lactotetra, a novel cell surface marker of undifferentiated human pluripotent stem cells. J Biol Chem 2014; 289:18846-59. [PMID: 24841197 PMCID: PMC4081926 DOI: 10.1074/jbc.m114.568832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cell surface glycoconjugates are used as markers for undifferentiated pluripotent stem cells. Here, antibody binding and mass spectrometry characterization of acid glycosphingolipids isolated from a large number (1 × 109 cells) of human embryonic stem cell (hESC) lines allowed identification of several novel acid glycosphingolipids, like the gangliosides sialyl-lactotetraosylceramide and sialyl-globotetraosylceramide, and the sulfated glycosphingolipids sulfatide, sulf-lactosylceramide, and sulf-globopentaosylceramide. A high cell surface expression of sialyl-lactotetra on hESC and human induced pluripotent stem cells (hiPSC) was demonstrated by flow cytometry, immunohistochemistry, and electron microscopy, whereas sulfated glycosphingolipids were only found in intracellular compartments. Immunohistochemistry showed distinct cell surface anti-sialyl-lactotetra staining on all seven hESC lines and three hiPSC lines analyzed, whereas no staining of hESC-derived hepatocyte-like or cardiomyocyte-like cells was obtained. Upon differentiation of hiPSC into hepatocyte-like cells, the sialyl-lactotetra epitope was rapidly down-regulated and not detectable after 14 days. These findings identify sialyl-lactotetra as a promising marker of undifferentiated human pluripotent stem cells.
Collapse
Affiliation(s)
- Angela Barone
- From the Institute of Clinical Sciences, Department of Surgery, S-41 345 Göteborg, Sweden
| | - Karin Säljö
- From the Institute of Clinical Sciences, Department of Surgery, S-41 345 Göteborg, Sweden
| | - John Benktander
- the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, S-40530 Göteborg, Sweden
| | - Maria Blomqvist
- the Institute of Biomedicine, Department of Clinical Chemistry and Transfusion Medicine, S-413 45 Göteborg, Sweden
| | - Jan-Eric Månsson
- the Institute of Biomedicine, Department of Clinical Chemistry and Transfusion Medicine, S-413 45 Göteborg, Sweden
| | - Bengt R Johansson
- the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, S-40530 Göteborg, Sweden
| | - Johan Mölne
- the Institute of Biomedicine, Department of Pathology, S-413 45 Göteborg, Sweden, and
| | - Anders Aspegren
- the Cellectis Stem Cells, Cellartis AB, S-413 46 Göteborg, Sweden
| | - Petter Björquist
- the Cellectis Stem Cells, Cellartis AB, S-413 46 Göteborg, Sweden
| | - Michael E Breimer
- From the Institute of Clinical Sciences, Department of Surgery, S-41 345 Göteborg, Sweden,
| | - Susann Teneberg
- the Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, S-40530 Göteborg, Sweden,
| |
Collapse
|
20
|
Skog M, Muhonen V, Nystedt J, Narcisi R, Kontturi LS, Urtti A, Korhonen M, van Osch GJVM, Kiviranta I. Xeno-free chondrogenesis of bone marrow mesenchymal stromal cells: towards clinical-grade chondrocyte production. Cytotechnology 2014; 67:905-19. [PMID: 24718835 DOI: 10.1007/s10616-014-9721-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 03/20/2014] [Indexed: 01/02/2023] Open
Abstract
Current cell-based cartilage therapies relay on articular cartilage-derived autologous chondrocytes as a cell source, which possesses disadvantages, such as, donor site morbidity and dedifferentiation of chondrocytes during in vitro expansion. Due to these and other limitations, novel cell sources and production strategies are needed. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are a fascinating alternative, but they are not spontaneously capable of producing hyaline cartilage-like repair tissue in vivo. In vitro pre-differentiation of BM-MSCs could be used to produce chondrocytes for clinical applications. However, clinically compatible defined and xeno-free differentiation protocol is lacking. Hence, this study aimed to develop such chondrogenic differentiation medium for human BM-MSCs. We assessed the feasibility of the medium using three human BM-MSCs donors and validated the method by comparing BM-MSCs to three other cell types holding potential for articular cartilage repair. The effectiveness of the method was compared to conventional serum-free and commercially available chondrogenic differentiation media. The results show that the defined xeno-free differentiation medium is at least as efficient as conventionally used serum-free chondrogenic medium and performed significantly better on all cell types tested compared to the commercially available chondrogenic medium.
Collapse
Affiliation(s)
- Maria Skog
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
ABCB1/MDR1 contributes to the anticancer drug-resistant phenotype of IPH-926 human lobular breast cancer cells. Cancer Lett 2011; 315:153-60. [PMID: 22118813 DOI: 10.1016/j.canlet.2011.09.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/22/2011] [Accepted: 09/27/2011] [Indexed: 11/21/2022]
Abstract
Contribution of the ABCB1/MDR1/P-glycoprotein drug transporter to breast cancer resistance has been controversial. One issue is that ABCB1-dependent drug-resistance has primarily been investigated in mammary epithelial cell models technically manipulated to overexpress ABCB1, either by gene transfer using appropriate expression vectors or by chronic anticancer drug-selection. However, an unmodified human breast cancer cell line with an endogenous overexpression of ABCB1 has not been described thus far. Using Affymetrix microarray analyses, we identified an endogenous overexpression of several tumor-biologically relevant transcripts including ABCB1, BCAR4, CCL28, SCGB2A2 and PIP in IPH-926, an anticancer drug-resistant human lobular breast cancer cell line derived from a chemo-refractory mammary carcinoma patient. In a panel of twenty breast cancer cell lines examined, overexpression of ABCB1 mRNA and protein was exclusively detected in IPH-926. This was further validated using chronically in vitro drug-selected KB-V-1 cells as a widely used reference model to accurately define an ABCB1 overexpression. IPH-926 and KB-V-1 displayed a similar overexpression of ABCB1. Flow cytometric analyses showed that IPH-926 but not ABCB1-negative breast cancer cells extruded the anticancer agent doxorubicin, a classical substrate of the ABCB1 drug transporter. PSC-833 (valspodar), a selective ABCB1 inhibitor, blocked this efflux, restored apoptotic PARP cleavage and increased doxorubicin sensitivity in IPH-926 and KB-V-1. To our knowledge, IPH-926 represents the first human breast cancer cell line with a genuine, endogenous overexpression of ABCB1. IPH-926 provides evidence that ABCB1 can occasionally cause anticancer drug-resistance in breast cancer patients and offers a new tool for the evaluation of compounds to overcome drug-resistance.
Collapse
|
22
|
GNE is involved in the early development of skeletal and cardiac muscle. PLoS One 2011; 6:e21389. [PMID: 21731727 PMCID: PMC3123316 DOI: 10.1371/journal.pone.0021389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/26/2011] [Indexed: 12/31/2022] Open
Abstract
UDP-N-acetylglucosamine 2 epimerase/N-acetylmannosamime kinase (GNE) is a bifunctional enzyme which catalyzes the two key sequential steps in the biosynthetic pathway of sialic acid, the most abundant terminal monosaccharide on glycoconjugates of eukaryotic cells. GNE knock out (GNE KO) mice are embryonically lethal at day E8.5. Although the role of GNE in the sialic pathway has been well established as well as the importance of sialylation in many diverse biological pathways, less is known about the involvement of GNE in muscle development. To address this issue we have studied the role of GNE during in vitro embryogenesis by comparing the developmental profile in culture of embryonic stem cells (ES) from wild type and from GNE KO E3.5 mice embryos, during 45 days. Neuronal cells appeared rarely in GNE KO ES cultures and did not reach an advanced differentiated stage. Although primary cardiac cells appeared at the same time in both normal and GNE KO ES cultures, GNE KO cardiac cells degraded very soon and their beating capacity decayed rapidly. Furthermore very rare skeletal muscle committed cells were detected in the GNE KO ES cultures at any stage of differentiation, as assessed by analysis of the expression of either Pax7, MyoD and MyHC markers. Beyond the supporting evidence that GNE plays an important role in neuronal cell and brain development, these results show that GNE is strongly involved in cardiac tissue and skeletal muscle early survival and organization. These findings could open new avenues in the understanding of muscle function mechanisms in health and in disease.
Collapse
|
23
|
Padler-Karavani V, Varki A. Potential impact of the non-human sialic acid N-glycolylneuraminic acid on transplant rejection risk. Xenotransplantation 2011; 18:1-5. [PMID: 21342282 PMCID: PMC3098739 DOI: 10.1111/j.1399-3089.2011.00622.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Vered Padler-Karavani
- Glycobiology Research and Training Center, Department of Medicine, University of California at San Diego, La Jolla, CA, USA.
| | | |
Collapse
|