51
|
Jaiswal M, Zhou M, Guo J, Tran TT, Kundu S, Jaufer AM, Fanucci GE, Guo Z. Different Biophysical Properties of Cell Surface α2,3- and α2,6-Sialoglycans Revealed by Electron Paramagnetic Resonance Spectroscopic Studies. J Phys Chem B 2023; 127:1749-1757. [PMID: 36808907 PMCID: PMC10116567 DOI: 10.1021/acs.jpcb.2c09048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Sialoglycans on HeLa cells were labeled with a nitroxide spin radical through enzymatic glycoengineering (EGE)-mediated installation of azide-modified sialic acid (Neu5Ac9N3) and then click reaction-based attachment of a nitroxide spin radical. α2,6-Sialyltransferase (ST) Pd2,6ST and α2,3-ST CSTII were used for EGE to install α2,6- and α2,3-linked Neu5Ac9N3, respectively. The spin-labeled cells were analyzed by X-band continuous wave (CW) electron paramagnetic resonance (EPR) spectroscopy to gain insights into the dynamics and organizations of cell surface α2,6- and α2,3-sialoglycans. Simulations of the EPR spectra revealed average fast- and intermediate-motion components for the spin radicals in both sialoglycans. However, α2,6- and α2,3-sialoglycans in HeLa cells possess different distributions of the two components, e.g., a higher average population of the intermediate-motion component for α2,6-sialoglycans (78%) than that for α2,3-sialoglycans (53%). Thus, the average mobility of spin radicals in α2,3-sialoglycans was higher than that in α2,6-sialoglycans. Given the fact that a spin-labeled sialic acid residue attached to the 6-O-position of galactose/N-acetyl-galactosamine would experience less steric hindrance and show more flexibility than that attached to the 3-O-position, these results may reflect the differences in local crowding/packing that restrict the spin-label and sialic acid motion for α2,6-linked sialoglycans. The studies further suggest that Pd2,6ST and CSTII may have different preferences for glycan substrates in the complex environment of the extracellular matrix. The discoveries of this work are biologically important as they are useful for interpreting the different functions of α2,6- and α2,3-sialoglycans and indicate the possibility of using Pd2,6ST and CSTII to target different glycoconjugates on cells.
Collapse
Affiliation(s)
- Mohit Jaiswal
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Mingwei Zhou
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Jiatong Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Trang T Tran
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Sayan Kundu
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Afnan M Jaufer
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Gail E Fanucci
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, Florida 32611, United States
| |
Collapse
|
52
|
Üstündağ FD, Ünal İ, Üstündağ ÜV, Cansız D, Beler M, Alturfan AA, Tiber PM, Emekli-Alturfan E. Morphine ameliorates pentylenetetrazole-induced locomotor pattern in zebrafish embryos; mechanism involving regulation of opioid receptors, suppression of oxidative stress, and inflammation in epileptogenesis. Toxicol Mech Methods 2023; 33:151-160. [PMID: 35866229 DOI: 10.1080/15376516.2022.2105182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Zebrafish (Danio rerio) is becoming an increasingly important model in epilepsy research. Pentylenetetrazole (PTZ) is a convulsant agent that induces epileptic seizure-like state in zebrafish and zebrafish embryos and is most commonly used in antiepileptic drug discovery research to evaluate seizure mechanisms. Classical antiepileptic drugs, such as valproic acid (VPA) reduce PTZ-induced epileptiform activities. Opioid system has been suggested to play a role in epileptogenesis. The aim of our study is to determine the effects of morphine in PTZ-induced epilepsy model in zebrafish embryos by evaluating locomotor activity and parameters related to oxidant-antioxidant status, inflammation, and cholinergic system as well as markers of neuronal activity c-fos, bdnf, and opioid receptors. Zebrafish embryos at 72 hpf were exposed to PTZ (20 mM), VPA (1 mM), and Morphine (MOR) (100 µM). MOR and VPA pretreated groups were treated with either MOR (MOR + PTZ) or VPA (VPA + PTZ) for 20 min before PTZ expoure. Locomotor activity was quantified as total distance moved (mm), average speed (mm/sec) and exploration rate (%) and analyzed using ToxTrac tracking programme. Oxidant-antioxidant system parameters, acetylcholinesterase activity, and sialic acid leves were evaluated using spectrophotometric methods. The expression of c-fos, bdnf, oprm1, and oprd1 were evaluated by RT-PCR. MOR pretreatment ameliorated PTZ-induced locomotor pattern as evidenced by improved average speed, exploration rate and distance traveled. We report the restoration of inflammatory and oxidant-antioxidant system parameters, c-fos, bdnf, and opioid receptor oprm1 as the possible mechanisms involved in the ameliorative effect of MOR against PTZ-induced epileptogenic process in zebrafish embryos.
Collapse
Affiliation(s)
- Fümet Duygu Üstündağ
- Department of Biophysics, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - İsmail Ünal
- Department of Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Ünsal Veli Üstündağ
- Faculty of Medicine, Medical Biochemistry, Istanbul Medipol University, Istanbul, Turkey
| | - Derya Cansız
- Faculty of Medicine, Medical Biochemistry, Istanbul Medipol University, Istanbul, Turkey
| | - Merih Beler
- Department of Biochemistry, Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - A Ata Alturfan
- Department of Biochemistry, Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Pınar Mega Tiber
- Department of Biophysics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Department of Basic Medical Sciences, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| |
Collapse
|
53
|
Abstract
Siglecs are a family of immunomodulatory cell surface receptors present on white blood cells. Binding to cell surface sialic acid-containing glycans modulates the proximity of Siglecs to other receptors that they regulate. This proximity is key to enabling signaling motifs on the cytosolic domain of Siglecs to modulate immune responses. Given the important roles that Siglecs play in helping to maintain immune homeostasis, a better understanding of their glycan ligands is needed to elucidate their roles in health and disease. A common approach for probing Siglec ligands on cells is to use soluble versions of the recombinant Siglecs in conjunction with flow cytometry. Flow cytometry has many advantages in enabling the relative levels of Siglec ligands between cell types to be rapidly quantified. Here, a step-by-step protocol is described on how to detect Siglec ligands most sensitively and accurately on cells by flow cytometry.
Collapse
Affiliation(s)
- Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
54
|
Saini P, Adeniji OS, Abdel-Mohsen M. Inhibitory Siglec-sialic acid interactions in balancing immunological activation and tolerance during viral infections. EBioMedicine 2022; 86:104354. [PMID: 36371982 PMCID: PMC9663867 DOI: 10.1016/j.ebiom.2022.104354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022] Open
Abstract
Siglecs are a family of emerging glyco-immune checkpoints. Inhibiting them can enhance the functions of several types of immune cells, whereas engaging them can reduce hyper-inflammation and hyper-activation of immune functions. Siglec-sialoglycan interactions play an important role in modulating immunological functions during cancer, however, their roles in regulating immunological equilibrium during viral infections is less clear. In this review, we discuss the documented and potential roles of inhibitory Siglecs in balancing immune activation and tolerance during viral infections and consider how this balance could affect both the desired anti-viral immunological functions and the unwanted hyper- or chronic inflammation. Finally, we discuss the opportunities to target the Siglec immunological switches to reach an immunological balance during viral infections: inhibiting specific Siglec-sialoglycan interactions when maximum anti-viral immune responses are needed, or inducing other interactions when preventing excessive inflammation or reducing chronic immune activation are the goals.
Collapse
|
55
|
Marciel MP, Haldar B, Hwang J, Bhalerao N, Bellis SL. Role of tumor cell sialylation in pancreatic cancer progression. Adv Cancer Res 2022; 157:123-155. [PMID: 36725107 PMCID: PMC11342334 DOI: 10.1016/bs.acr.2022.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies and is currently the third leading cause of cancer death. The aggressiveness of PDAC stems from late diagnosis, early metastasis, and poor efficacy of current chemotherapies. Thus, there is an urgent need for effective biomarkers for early detection of PDAC and development of new therapeutic strategies. It has long been known that cellular glycosylation is dysregulated in pancreatic cancer cells, however, tumor-associated glycans and their cognate glycosylating enzymes have received insufficient attention as potential clinical targets. Aberrant glycosylation affects a broad range of pathways that underpin tumor initiation, metastatic progression, and resistance to cancer treatment. One of the prevalent alterations in the cancer glycome is an enrichment in a select group of sialylated glycans including sialylated, branched N-glycans, sialyl Lewis antigens, and sialylated forms of truncated O-glycans such as the sialyl Tn antigen. These modifications affect the activity of numerous cell surface receptors, which collectively impart malignant characteristics typified by enhanced cell proliferation, migration, invasion and apoptosis-resistance. Additionally, sialic acids on tumor cells engage inhibitory Siglec receptors on immune cells to dampen anti-tumor immunity, further promoting cancer progression. The goal of this review is to summarize the predominant changes in sialylation occurring in pancreatic cancer, the biological functions of sialylated glycoproteins in cancer pathogenesis, and the emerging strategies for targeting sialoglycans and Siglec receptors in cancer therapeutics.
Collapse
Affiliation(s)
- Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Barnita Haldar
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nikita Bhalerao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
56
|
Wang JL, Hu XY, Han CG, Hou SY, Wang HS, Zheng F. Lanthanide Complexes for Tumor Diagnosis and Therapy by Targeting Sialic Acid. ACS NANO 2022; 16:14827-14837. [PMID: 35981089 DOI: 10.1021/acsnano.2c05715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Sialic acid (SA) is overexpressed on cell membranes of tumor cells, and increased serum SA concentration has been observed in tumor-bearing patients. Herein, a series of lanthanide-containing bimetallic complexes (TDA-M-Lns) for targeting SA were prepared via coordination among luminescent lanthanide ions (Ln3+ = Tb3+, Eu3+, Dy3+, or Sm3+), metal ion quenchers (M2+ = Cu2+ or Co2+), and the organic ligand 2,2'-thiodiacetic acid (TDA). SA can competitively coordinate with Ln3+, resulting in the "signal-on" of the Ln3+. Therefore, the TDA-M-Lns can be simply used for cost-saving detection of SA in the blood samples. Among the TDA-M-Lns, TDA-Co-Eu showed the highest sensitivity to detect SA in the blood of tumor-bearing mice. Furthermore, the TDA-Co-Eu was successfully used to target SA and deposit Eu3+ on the surfaces of tumor cells for the inhibition of tumor cell growth and migration. The therapeutic effect of TDA-Co-Eu on a Balb/c mouse liver tumor model was evaluated. It was proved that TDA-Co-Eu can be applied for SA detection as well as for inhibiting tumor growth.
Collapse
Affiliation(s)
- Jia-Li Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Xin-Yuan Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng-Gang Han
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Shao-Yuan Hou
- Administration for Market Regulation of Shanting district, Zaozhuang 277200, China
| | - Huai-Song Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Zheng
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
57
|
Wen R, Zhao H, Zhang D, Chiu CL, Brooks JD. Sialylated glycoproteins as biomarkers and drivers of progression in prostate cancer. Carbohydr Res 2022; 519:108598. [PMID: 35691122 DOI: 10.1016/j.carres.2022.108598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023]
Abstract
Sialic acids have been implicated in cancer initiation, progression, and immune evasion in diverse human malignancies. Sialylation of terminal glycans on cell surface and secreted glycoproteins is a long-recognized feature of cancer cells. Recently, immune checkpoint inhibitor immunotherapy has tremendously improved the outcomes of patients with various cancers. However, available immunotherapy approaches have had limited efficacy in metastatic castration-resistant prostate cancer. Sialic acid modified glycoproteins in prostate cancers and their interaction with Siglec receptors on tumor infiltrating immune cells might underlie immunosuppressive signaling in prostate cancer. Here, we summarize the function of sialic acids and relevant glycosynthetic enzymes in cancer initiation and progression. We also discuss the possible uses of sialic acids as biomarkers in prostate cancer and the potential methods for targeting Siglec-sialic acid interactions for prostate cancer treatment.
Collapse
Affiliation(s)
- Ru Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
58
|
Aberrant Sialylation in Cancer: Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14174248. [PMID: 36077781 PMCID: PMC9454432 DOI: 10.3390/cancers14174248] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
The surface of every eukaryotic cell is coated in a thick layer of glycans that acts as a key interface with the extracellular environment. Cancer cells have a different ‘glycan coat’ to healthy cells and aberrant glycosylation is a universal feature of cancer cells linked to all of the cancer hallmarks. This means glycans hold huge potential for the development of new diagnostic and therapeutic strategies. One key change in tumour glycosylation is increased sialylation, both on N-glycans and O-glycans, which leads to a dense forest of sialylated structures covering the cell surface. This hypersialylation has far-reaching consequences for cancer cells, and sialylated glycans are fundamental in tumour growth, metastasis, immune evasion and drug resistance. The development of strategies to inhibit aberrant sialylation in cancer represents an important opportunity to develop new therapeutics. Here, I summarise recent advances to target aberrant sialylation in cancer, including the development of sialyltransferase inhibitors and strategies to inhibit Siglecs and Selectins, and discuss opportunities for the future.
Collapse
|
59
|
Abuduxikuer K, Wang L, Zou L, Cao CY, Yu L, Guo HM, Liang XM, Wang JS, Chen L. Updated clinical and glycomic features of mannosyl-oligosaccharide glucosidase deficiency: Two case reports. World J Clin Cases 2022; 10:7397-7408. [PMID: 36158009 PMCID: PMC9353930 DOI: 10.12998/wjcc.v10.i21.7397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/16/2021] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mannosyl-oligosaccharide glucosidase (MOGS) deficiency is an extremely rare type of congenital disorder of glycosylation (CDG), with only 12 reported cases. Its clinical, genetic, and glycomic features are still expanding. Our aim is to update the novel clinical and glycosylation features of 2 previously reported patients with MOGS-CDG.
CASE SUMMARY We collected comprehensive clinical information, and conducted the immunoglobulin G1 glycosylation assay using nano-electrospray ionization source quadruple time-of-flight mass spectrometry. Novel dysmorphic features included an enlarged tongue, forwardly rotated earlobes, a birth mark, overlapped toes, and abnormal fat distribution. Novel imaging findings included pericardial effusion, a deep interarytenoid groove, mild congenital subglottic stenosis, and laryngomalacia. Novel laboratory findings included peripheral leukocytosis with neutrophil predominance, elevated C-reactive protein and creatine kinase, dyslipidemia, coagulopathy, complement 3 and complement 4 deficiencies, decreased proportions of T lymphocytes and natural killer cells, and increased serum interleukin 6. Glycosylation studies showed a significant increase of hypermannosylated glycopeptides (Glc3Man7GlcNAc2/N2H10 and Man5GlcNAc2/N2H5) and hypersialylated glycopeptides. A compensatory glycosylation pathway leading to an increase in Man5GlcNAc2/N2H5 was indicated with the glycosylation profile.
CONCLUSION We confirmed abnormal glycomics in 1 patient, expanding the clinical and glycomic spectrum of MOGS-CDG. We also postulated a compensatory glycosylation pathway, leading to a possible serum biomarker for future diagnosis.
Collapse
Affiliation(s)
| | - Lei Wang
- Department of Research and Development, SysDiagno Biomedtech, Nanjing 211800, Jiangsu Province, China
| | - Lin Zou
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Cui-Yan Cao
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Long Yu
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Hong-Mei Guo
- Department of Gastroenterology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xin-Miao Liang
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Jian-She Wang
- Department of Hepatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Li Chen
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
60
|
Moshe Halamish H, Zlotver I, Sosnik A. Polymeric nanoparticles surface-complexed with boric acid actively target solid tumors overexpressing sialic acid. J Colloid Interface Sci 2022; 626:916-929. [PMID: 35835042 DOI: 10.1016/j.jcis.2022.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022]
Abstract
Sialic acid is a fundamental component of the tumor microenvironment, modulates cell-cell and cell-extracellular matrix interactions and is associated with bad prognosis and clinical outcomes in different cancers. Capitalizing on the ability of boric acid to form cyclic esters with diols, in this work, we design self-assembled multi-micellar colloidal systems of an amphiphilic poly(vinyl alcohol)-g-poly(methyl methacrylate) copolymer surface-modified with boric acid for the active targeting of solid tumors that overexpress sialic acid. Nanoparticles display sizes in the 100-200 nm range and a spherical morphology, as determined by dynamic light scattering and high resolution-scanning electron microscopy, respectively. The uptake and anti-proliferative activity are assessed in 2D and 3D models of rhabdomyosarcoma in vitro. Surface boration increases the nanoparticle permeability and uptake, especially in rhabdomyosarcoma spheroids that overexpress sialic acid to a greater extent than 2D cultures. The biodistribution of non-borated and borated nanoparticles upon intravenous injection to a subcutaneous rhabdomyosarcoma murine xenograft model confirm a statistically significant increase in the intertumoral accumulation of the modified nanocarriers with respect to the unmodified counterparts and a sharp decrease in major clearance organs such as the liver. Overall, our results highlight the promise of these borated nanomaterials to actively target hypersialylated solid tumors.
Collapse
Affiliation(s)
- Hen Moshe Halamish
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel
| | - Ivan Zlotver
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel.
| |
Collapse
|
61
|
Burge K, Eckert J, Wilson A, Trammell M, Lueschow SR, McElroy SJ, Dyer D, Chaaban H. Hyaluronic Acid 35 kDa Protects against a Hyperosmotic, Formula Feeding Model of Necrotizing Enterocolitis. Nutrients 2022; 14:nu14091779. [PMID: 35565748 PMCID: PMC9105773 DOI: 10.3390/nu14091779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/23/2022] Open
Abstract
Necrotizing enterocolitis (NEC), an inflammatory disease of the intestine, is a common gastrointestinal emergency among preterm infants. Intestinal barrier dysfunction, hyperactivation of the premature immune system, and dysbiosis are thought to play major roles in the disease. Human milk (HM) is protective, but the mechanisms underpinning formula feeding as a risk factor in the development of NEC are incompletely understood. Hyaluronic acid 35 kDa (HA35), a bioactive glycosaminoglycan of HM, accelerates intestinal development in murine pups during homeostasis. In addition, HA35 prevents inflammation-induced tissue damage in pups subjected to murine NEC, incorporating Paneth cell dysfunction and dysbiosis. We hypothesized HA35 treatment would reduce histological injury and mortality in a secondary mouse model of NEC incorporating formula feeding. NEC-like injury was induced in 14-day mice by dithizone-induced disruption of Paneth cells and oral gavage of rodent milk substitute. Mortality and histological injury, serum and tissue cytokine levels, stool bacterial sequencing, and bulk RNA-Seq comparisons were analyzed. HA35 significantly reduced the severity of illness in this model, with a trend toward reduced mortality, while RNA-Seq analysis demonstrated HA35 upregulated genes associated with goblet cell function and innate immunity. Activation of these critical protective and reparative mechanisms of the small intestine likely play a role in the reduced pathology and enhanced survival trends of HA-treated pups subjected to intestinal inflammation in this secondary model of NEC, providing potentially interesting translational targets for the human preterm disease.
Collapse
Affiliation(s)
- Kathryn Burge
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - Jeffrey Eckert
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - Adam Wilson
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Shiloh R. Lueschow
- Department of Microbiology and Immunology, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven J. McElroy
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA;
| | - David Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (M.T.); (D.D.)
| | - Hala Chaaban
- Department of Pediatrics, Division of Neonatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.B.); (J.E.); (A.W.)
- Correspondence:
| |
Collapse
|
62
|
Sialic acids: An Avenue to Target Cancer Progression, Metastasis, and Resistance to Therapy. FORUM OF CLINICAL ONCOLOGY 2022. [DOI: 10.2478/fco-2021-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Abstract
Background
Sialic acids are alpha-keto acids with nine carbons that are commonly present in the terminal sugars of glycans on glycoproteins and glycolipids on the cell surface. Sialic acids have a role in a variety of physiological and pathological processes by interacting with carbohydrates and proteins, communicating between cells, and acting as cell surface receptors for viruses and bacteria. Several studies have shown the aberrant pattern of sialic acids on cancer cells due to change in their glycosylation status. This pattern may be attributed to various physiological and pathological changes occurring in tumour cells. Hypersialylation in tumours, its involvement in tumour growth, immune evasion and escape from the apoptotic pathway, metastasis formation, and therapeutic resistance have all been fairly well investigated.
Methods
A PubMed search was conducted and published articles in different studies from 2000 to 2020 were included and reviewed. Here, we discuss current outcomes that emphasize the unfavourable effects of hypersialylation on multiple aspects of tumour genesis, immune evasion, metastasis and resistance to therapy.
Conclusion
These recent investigations have found that aberrant sialylation is an essential process for tumour cells to evade immune surveillance and maintain their malignancy. Together, these noteworthy views provide a solid platform for designing and developing therapeutic approaches that target hypersialylation of cancer cells.
Collapse
|
63
|
Beyer S, Kimani M, Zhang Y, Verhassel A, Sternbæk L, Wang T, Persson JL, Härkönen P, Johansson E, Caraballo R, Elofsson M, Gawlitza K, Rurack K, Ohlsson L, El-Schich Z, Wingren AG, Stollenwerk MM. Fluorescent Molecularly Imprinted Polymer Layers against Sialic Acid on Silica-Coated Polystyrene Cores—Assessment of the Binding Behavior to Cancer Cells. Cancers (Basel) 2022; 14:cancers14081875. [PMID: 35454783 PMCID: PMC9024825 DOI: 10.3390/cancers14081875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/21/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Sialic acid (SA) is a monosaccharide usually linked to the terminus of glycan chains on the cell surface. It plays a crucial role in many biological processes, and hypersialylation is a common feature in cancer. Lectins are widely used to analyze the cell surface expression of SA. However, these protein molecules are usually expensive and easily denatured, which calls for the development of alternative glycan-specific receptors and cell imaging technologies. In this study, SA-imprinted fluorescent core-shell molecularly imprinted polymer particles (SA-MIPs) were employed to recognize SA on the cell surface of cancer cell lines. The SA-MIPs improved suspensibility and scattering properties compared with previously used core-shell SA-MIPs. Although SA-imprinting was performed using SA without preference for the α2,3- and α2,6-SA forms, we screened the cancer cell lines analyzed using the lectins Maackia Amurensis Lectin I (MAL I, α2,3-SA) and Sambucus Nigra Lectin (SNA, α2,6-SA). Our results show that the selected cancer cell lines in this study presented a varied binding behavior with the SA-MIPs. The binding pattern of the lectins was also demonstrated. Moreover, two different pentavalent SA conjugates were used to inhibit the binding of the SA-MIPs to breast, skin, and lung cancer cell lines, demonstrating the specificity of the SA-MIPs in both flow cytometry and confocal fluorescence microscopy. We concluded that the synthesized SA-MIPs might be a powerful future tool in the diagnostic analysis of various cancer cells.
Collapse
Affiliation(s)
- Sarah Beyer
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
| | - Martha Kimani
- Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter Straße 11, 12489 Berlin, Germany; (M.K.); (K.G.); (K.R.)
| | - Yuecheng Zhang
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
| | - Alejandra Verhassel
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.V.); (P.H.)
- FICAN West Cancer Centre, Turku University Hospital, 20520 Turku, Finland
| | - Louise Sternbæk
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
- Phase Holographic Imaging AB, SE-223 63 Lund, Sweden
| | - Tianyan Wang
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden;
| | - Jenny L. Persson
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden;
| | - Pirkko Härkönen
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland; (A.V.); (P.H.)
- FICAN West Cancer Centre, Turku University Hospital, 20520 Turku, Finland
| | - Emil Johansson
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden; (E.J.); (R.C.); (M.E.)
- Umeå Centre for Microbial Research, Umeå University, SE-901 87 Umeå, Sweden
| | - Remi Caraballo
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden; (E.J.); (R.C.); (M.E.)
- Umeå Centre for Microbial Research, Umeå University, SE-901 87 Umeå, Sweden
| | - Mikael Elofsson
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden; (E.J.); (R.C.); (M.E.)
- Umeå Centre for Microbial Research, Umeå University, SE-901 87 Umeå, Sweden
| | - Kornelia Gawlitza
- Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter Straße 11, 12489 Berlin, Germany; (M.K.); (K.G.); (K.R.)
| | - Knut Rurack
- Chemical and Optical Sensing Division, Bundesanstalt für Materialforschung und -prüfung (BAM), Richard-Willstätter Straße 11, 12489 Berlin, Germany; (M.K.); (K.G.); (K.R.)
| | - Lars Ohlsson
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
| | - Zahra El-Schich
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
- Correspondence:
| | - Maria M. Stollenwerk
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, SE-205 06 Malmö, Sweden; (S.B.); (Y.Z.); (L.S.); (J.L.P.); (L.O.); (Z.E.-S.); (M.M.S.)
- Biofilms-Research Center for Biointerfaces, Malmö University, SE-205 06 Malmö, Sweden
| |
Collapse
|
64
|
Ko CY, Chu TH, Hsu CC, Chen HP, Huang SC, Chang CL, Tzou SJ, Chen TY, Lin CC, Shih PC, Lin CH, Chang CF, Lee YK. Bioinformatics Analyses Identify the Therapeutic Potential of ST8SIA6 for Colon Cancer. J Pers Med 2022; 12:jpm12030401. [PMID: 35330401 PMCID: PMC8953768 DOI: 10.3390/jpm12030401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/27/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Sialylation of glycoproteins is modified by distinct sialyltransferases such as ST3Gal, ST6Gal, ST6GalNAc, or ST8SIA with α2,3-, α2,6-, or α2,8-linkages. Alteration of these sialyltransferases causing aberrant sialylation is associated with the progression of colon cancer. However, among the ST8- sialyltransferases, the role of ST8SIA6 in colon cancer remains poorly understood. In this study, we explored the involvement of ST8SIA6 in colon cancer using multiple gene databases. The relationship between ST8SIA6 expression and tumor stages/grades was investigated by UALCAN analysis, and Kaplan–Meier Plotter analysis was used to analyze the expression of ST8SIA6 on the survival outcome of colon cancer patients. Moreover, the biological functions of ST8SIA6 in colon cancer were explored using LinkedOmics and cancer cell metabolism gene DB. Finally, TIMER and TISMO analyses were used to delineate ST8SIA6 levels in tumor immunity and immunotherapy responses, respectively. ST8SIA6 downregulation was associated with an advanced stage and poorly differentiated grade; however, ST8SIA6 expression did not affect the survival outcomes in patients with colon cancer. Gene ontology analysis suggested that ST8SIA6 participates in cell surface adhesion, angiogenesis, and membrane vesicle trafficking. In addition, ST8SIA6 levels affected immunocyte infiltration and immunotherapy responses in colon cancer. Collectively, these results suggest that ST8SIA6 may serve as a novel therapeutic target towards personalized medicine for colon cancer.
Collapse
Affiliation(s)
- Chou-Yuan Ko
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-C.H.); (C.-L.C.); (S.-J.T.)
| | - Tian-Huei Chu
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Ching-Cheng Hsu
- Department of Internal Medicine, Division of Cardiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (P.-C.S.); (C.-H.L.)
| | - Hsin-Pao Chen
- Department of Surgery, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan;
| | - Shih-Chung Huang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-C.H.); (C.-L.C.); (S.-J.T.)
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Division of Cardiology, Department of Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Chen-Lin Chang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-C.H.); (C.-L.C.); (S.-J.T.)
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Shiow-Jyu Tzou
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-C.H.); (C.-L.C.); (S.-J.T.)
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Department of Nursing, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Tung-Yuan Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
| | - Chia-Chen Lin
- Clinical Pathology Department, Chi Mei Medical Center, Tainan 71004, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pei-Chun Shih
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (P.-C.S.); (C.-H.L.)
| | - Chung-Hsien Lin
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (P.-C.S.); (C.-H.L.)
| | - Chuan-Fa Chang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; (P.-C.S.); (C.-H.L.)
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Correspondence: (C.-F.C.); (Y.-K.L.); Tel.: +886-6-235-3535 (ext. 5796) (C.-F.C.); +886-7-749-6751 (ext. 726201) (Y.-K.L.)
| | - Yung-Kuo Lee
- Medical Laboratory, Medical Education and Research Center, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan;
- Correspondence: (C.-F.C.); (Y.-K.L.); Tel.: +886-6-235-3535 (ext. 5796) (C.-F.C.); +886-7-749-6751 (ext. 726201) (Y.-K.L.)
| |
Collapse
|
65
|
Bindeman WE, Fingleton B. Glycosylation as a regulator of site-specific metastasis. Cancer Metastasis Rev 2022; 41:107-129. [PMID: 34967926 PMCID: PMC8930623 DOI: 10.1007/s10555-021-10015-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
Metastasis is considered to be responsible for 90% of cancer-related deaths. Although it is clinically evident that metastatic patterns vary by primary tumor type, the molecular mechanisms underlying the site-specific nature of metastasis are an area of active investigation. One mechanism that has emerged as an important player in this process is glycosylation, or the addition of sugar moieties onto protein and lipid substrates. Glycosylation is the most common post-translational modification, occurring on more than 50% of translated proteins. Many of those proteins are either secreted or expressed on the cell membrane, thereby making glycosylation an important mediator of cell-cell interactions, including tumor-microenvironment interactions. It has been recently discovered that alteration of glycosylation patterns influences cancer metastasis, both globally and in a site-specific manner. This review will summarize the current knowledge regarding the role of glycosylation in the tropism of cancer cells for several common metastatic sites, including the bone, lung, brain, and lymph nodes.
Collapse
Affiliation(s)
- Wendy E Bindeman
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Barbara Fingleton
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
66
|
Abstract
CD47 is a "don't eat me" signal to phagocytes that is overexpressed on many tumor cells as a potential mechanism for immune surveillance evasion. CD47 and its interaction with signal-regulating protein alpha (SIRPα) on phagocytes is therefore a promising cancer target. Therapeutic antibodies and fusion proteins that block CD47 or SIRPα have been developed and have shown activity in preclinical models of hematologic and solid tumors. Anemia is a common adverse event associated with anti-CD47 treatment, but mitigation strategies-including use of a low 'priming' dose-have substantially reduced this risk in clinical studies. While efficacy in single-agent clinical studies is lacking, findings from studies of CD47-SIRPα blockade in combination with agents that increase 'eat me' signals or with antitumor antibodies are promising. Magrolimab, an anti-CD47 antibody, is the furthest along in clinical development among agents in this class. Magrolimab combination therapy in phase Ib/II studies has been well tolerated with encouraging response rates in hematologic and solid malignancies. Similar combination therapy studies with other anti-CD47-SIRPα agents are beginning to report. Based on these early clinical successes, many trials have been initiated in hematologic and solid tumors testing combinations of CD47-SIRPα blockade with standard therapies. The results of these studies will help determine the role of this novel approach in clinical practice and are eagerly awaited.
Collapse
Affiliation(s)
- R. Maute
- Gilead Sciences, Inc., Foster City, USA
| | - J. Xu
- Gilead Sciences, Inc., Foster City, USA
| | - I.L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
67
|
Sgambati E, Tani A, Leri M, Delfino G, Zecchi-Orlandini S, Bucciantini M, Nosi D. Correlation between Sialylation Status and Cell Susceptibility to Amyloid Toxicity. Cells 2022; 11:cells11040601. [PMID: 35203252 PMCID: PMC8870280 DOI: 10.3390/cells11040601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023] Open
Abstract
The interaction between the cell membrane and misfolded protein species plays a crucial role in the development of neurodegeneration. This study was designed to clarify the relationship between plasma membrane composition in terms of the differently linked sialic acid (Sia) content and cell susceptibility to toxic and misfolded Aβ-42 peptides. The sialylation status in different cell lines was investigated by lectin histochemistry and confocal immunofluorescence and then correlated with the different propensities to bind amyloid fibrils and with the relative cell susceptibility to amyloid damage. This study reveals that expressions of Sias α2,3 and α2,6 linked to galactose/N-acetyl-galactosamine, and PolySia are positively correlated with Aβ-42-induced cell toxicity. PolySia shows an early strong interaction with amyloid fibrils, favoring their binding to GM1 ganglioside containing α2,3 galactose-linked Sia and a loss of cell viability. Our findings demonstrate that cell lines with a prevailing plastic neuron-like phenotype and high monoSia and PolySia contents are highly susceptible to amyloid Aβ-42 toxicity. This toxicity may involve a change in neuron metabolism and promote a compensative/protective increase in PolySia, which, in turn, could favor amyloid binding to GM1, thus exacerbating cell dysmetabolism and further amyloid aggregation.
Collapse
Affiliation(s)
- Eleonora Sgambati
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy;
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (A.T.); (S.Z.-O.); (D.N.)
| | - Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
| | - Giovanni Delfino
- Department of Biology (BIO), University of Florence, Via Giorgio La Pira 4, 50121 Florence, Italy;
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (A.T.); (S.Z.-O.); (D.N.)
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy;
- Correspondence:
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (A.T.); (S.Z.-O.); (D.N.)
| |
Collapse
|
68
|
Dong H, Gao Y, Huang X, Wu X. Synthesis of sialic acid conjugates of the clinical near-infrared dye as next-generation theranostics for cancer phototherapy. J Mater Chem B 2022; 10:927-934. [PMID: 35060591 PMCID: PMC9112073 DOI: 10.1039/d1tb02693c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cancer is a multifaceted global health problem that requires continuous action to develop next-generation cancer theranostics. Inspired by the emerging use of indocyanine green (ICG), the only clinically approved near-infrared (NIR) dye for cancer phototherapy, here we synthesized two ICG conjugate theranostics by coupling ICG to sialic acid (Sia) through the C2 and C9 positions of Sia, respectively, referred to as Sia-C2-ICG and Sia-C9-ICG. Encouragingly, Sia-C2/C9-ICGs show superior in vitro properties, including enhanced stability, reduced non-specific binding to serum proteins, and improved blood compatibility, highlighting the benefits of Sia coupling. Notably, in vivo NIR imaging shows that Sia-C9-ICG significantly promotes tumor targeting and effectively prolongs the circulation time in the body, while Sia-C2-ICG is superior to ICG but inferior to Sia-C9-ICG in targeting tumors. Furthermore, Sia-C9-ICG combined with NIR laser irradiation can lead to excellent photothermal and photodynamic therapies for cancer cells, resulting in superior solid tumor ablation. To our knowledge, this is the first report of Sia-NIR conjugates achieving significant tumor reduction in vivo. Together, these advances render Sia-C9-ICG an attractive lead as next-generation cancer theranostics that can be translated clinically to treat human patients.
Collapse
Affiliation(s)
- Huiling Dong
- National Glycoengineering Research Center, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong 266237, China.
| | - Yanan Gao
- National Glycoengineering Research Center, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong 266237, China.
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuanjun Wu
- National Glycoengineering Research Center, and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, Shandong 266237, China.
- Suzhou Research Institute, Shandong University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
69
|
Kim HJ, Schweiker S, Powell K, Levonis S. An efficient and robust HPLC method to determine the sialylation levels of human epithelial cells. PLoS One 2022; 17:e0257178. [PMID: 35041670 PMCID: PMC8765648 DOI: 10.1371/journal.pone.0257178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
Sialyltransferase, an enzyme responsible for attaching sialic acid to the cell surface, is reported to play a key role in cancer, making sialyltransferase a potential therapeutic target in drug development. Several methods have been developed to quantify sialic acids in biological samples however limitations exists and quantification in complex cell matrices lack investigation. Hence, this paper outlines a simple method to detect and quantify sialic acids in cancer cells for evaluating sialyltransferase activity of potential therapeutic compounds. An efficient method was developed using a reverse-phase ion-pairing HPLC-UV using triisopropanolamine as the ion-pairing agent with a C18 column. Neu5Ac was successfully eluted with the retention time 6.344 min with a flow rate of 0.4 mL/min. The proposed method was validated appropriately according to the AOAC guidelines (2013). This work demonstrates that the proposed method is not only relatively simple but also cost and time effective compared to pre-existing methods to successfully determine both free and protein-bound Neu5Ac in a complex cancer cell matrix. Furthermore, by applying the proposed method, a statistically significant decrease was observed for both HeLa and HuCCT1 cell lines with the application of deoxycholic acid-a known sialyltransferase inhibitor. Hence, the proposed method seems promisingly applicable to evaluate the effectiveness of potential sialyltransferase inhibitors.
Collapse
Affiliation(s)
- Hyo Jeong Kim
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Stephanie Schweiker
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Katie Powell
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Stephan Levonis
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland, Australia
| |
Collapse
|
70
|
Hait NC, Maiti A, Wu R, Andersen VL, Hsu CC, Wu Y, Chapla DG, Takabe K, Rusiniak ME, Bshara W, Zhang J, Moremen KW, Lau JTY. Extracellular sialyltransferase st6gal1 in breast tumor cell growth and invasiveness. Cancer Gene Ther 2022; 29:1662-1675. [PMID: 35676533 PMCID: PMC9663294 DOI: 10.1038/s41417-022-00485-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/09/2022] [Accepted: 05/20/2022] [Indexed: 02/04/2023]
Abstract
The sialyltransferase ST6GAL1 that adds α2-6 linked sialic acids to N-glycans of cell surface and secreted glycoproteins is prominently associated with many human cancers. Tumor-native ST6GAL1 promotes tumor cell behaviors such as invasion and resistance to cell stress and chemo- and radio-treatments. Canonically, ST6GAL1 resides in the intracellular secretory apparatus and glycosylates nascent glycoproteins in biosynthetic transit. However, ST6GAL1 is also released into the extracellular milieu and extracellularly remodels cell surface and secreted glycans. The impact of this non-canonical extrinsic mechanism of ST6GAL1 on tumor cell pathobiology is not known. We hypothesize that ST6GAL1 action is the combined effect of natively expressed sialyltransferase acting cell-autonomously within the ER-Golgi complex and sialyltransferase from extracellular origins acting extrinsically to remodel cell-surface glycans. We found that shRNA knockdown of intrinsic ST6GAL1 expression resulted in decreased ST6GAL1 cargo in the exosome-like vesicles as well as decreased breast tumor cell growth and invasive behavior in 3D in vitro cultures. Extracellular ST6GAL1, present in cancer exosomes or the freely soluble recombinant sialyltransferase, compensates for insufficient intrinsic ST6GAL1 by boosting cancer cell proliferation and increasing invasiveness. Moreover, we present evidence supporting the existence novel but yet uncharacterized cofactors in the exosome-like particles that potently amplify extrinsic ST6GAL1 action, highlighting a previously unknown mechanism linking this enzyme and cancer pathobiology. Our data indicate that extracellular ST6GAL1 from remote sources can compensate for cellular ST6GAL1-mediated aggressive tumor cell proliferation and invasive behavior and has great clinical potential for extracellular ST6GAL1 as these molecules are in the extracellular space should be easily accessible targets.
Collapse
Affiliation(s)
- Nitai C. Hait
- grid.240614.50000 0001 2181 8635Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA ,grid.240614.50000 0001 2181 8635Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Aparna Maiti
- grid.240614.50000 0001 2181 8635Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA ,grid.240614.50000 0001 2181 8635Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Rongrong Wu
- grid.240614.50000 0001 2181 8635Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Valerie L. Andersen
- grid.240614.50000 0001 2181 8635Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Chang-Chieh Hsu
- grid.273335.30000 0004 1936 9887Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260 USA
| | - Yun Wu
- grid.273335.30000 0004 1936 9887Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260 USA
| | - Digantkumar G. Chapla
- grid.213876.90000 0004 1936 738XComplex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA ,grid.213876.90000 0004 1936 738XDepartment of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA
| | - Kazuaki Takabe
- grid.240614.50000 0001 2181 8635Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Michael E. Rusiniak
- grid.240614.50000 0001 2181 8635Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Wiam Bshara
- grid.240614.50000 0001 2181 8635Department of Pathology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263 USA
| | - Jianmin Zhang
- grid.240614.50000 0001 2181 8635Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14203 USA
| | - Kelley W. Moremen
- grid.213876.90000 0004 1936 738XComplex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA ,grid.213876.90000 0004 1936 738XDepartment of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA
| | - Joseph T. Y. Lau
- grid.240614.50000 0001 2181 8635Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| |
Collapse
|
71
|
Rosa-Fernandes L, Oba-Shinjo SM, Macedo-da-Silva J, Marie SKN, Palmisano G. Aberrant Protein Glycosylation in Brain Cancers, with Emphasis on Glioblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:39-70. [DOI: 10.1007/978-3-031-05460-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
72
|
Choi H, Ho M, Adeniji OS, Giron L, Bordoloi D, Kulkarni AJ, Puchalt AP, Abdel-Mohsen M, Muthumani K. Development of Siglec-9 Blocking Antibody to Enhance Anti-Tumor Immunity. Front Oncol 2021; 11:778989. [PMID: 34869028 PMCID: PMC8640189 DOI: 10.3389/fonc.2021.778989] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Sialic acid-binding Immunoglobulin-like lectin-9 (Siglec-9) is a glyco-immune negative checkpoint expressed on several immune cells. Siglec-9 exerts its inhibitory effects by binding to sialoglycan ligands expressed on cancer cells, enabling them to evade immunosurveillance. We developed a panel of human anti-Siglec-9 hybridoma clones by immunizing mice with Siglec-9-encoding DNA and Siglec-9 protein. The lead antibodies, with high specificity and functionality against Siglec-9, were identified through screening of clones. The in vitro cytotoxicity assays showed that our lead antibody enhances anti-tumor immune activity. Further, in vivo testing utilizing ovarian cancer humanized mouse model showed a drastic reduction in tumor volume. Together, we developed novel antibodies that augment anti-tumor immunity through interference with Siglec-9-mediated immunosuppression.
Collapse
Affiliation(s)
- Hyeree Choi
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Michelle Ho
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Opeyemi S Adeniji
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Leila Giron
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Devivasha Bordoloi
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Abhijeet J Kulkarni
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | | | - Mohamed Abdel-Mohsen
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| | - Kar Muthumani
- Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
73
|
Rana R, Rani S, Kumar V, Nakhate KT, Ajazuddin, Gupta U. Sialic Acid Conjugated Chitosan Nanoparticles: Modulation to Target Tumour Cells and Therapeutic Opportunities. AAPS PharmSciTech 2021; 23:10. [PMID: 34862568 DOI: 10.1208/s12249-021-02170-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Targeted delivery of therapeutics forestalls the dreadful delocalized effects, drug toxicities and needless immunosuppression. Cancer cells are bounteous with sialic acid and the differential expression of glycosyl transferase, glycosidase and monosaccharide transporter compared to healthy tissues. The current study entails the development and characterisation of sialic acid (SA)-labelled chitosan nanoparticles encapsulating gemcitabine (GEM). Chitosan (CS) was conjugated with SA using coupling reaction and characterised spectroscopically. Furthermore, different concentrations of chitosan and tripolyphosphate (TPP) were optimised to fabricate surface modified chitosan nanoparticles. SA conjugated chitosan nanoparticles encapsulating GEM (SA-CS_GEM NPs) of 232 ± 9.69 nm with narrow distribution (PDI < 0.5) and zeta potential of - 19 ± 0.97 mV was fabricated. GEM was successfully loaded in the SA-CS NPs, depicting prolonged and biphasic drug release pattern more elated at low pH. Pronounced cellular uptake (FITC tagged) and cytotoxicity (IC50 487.4 nM) was observed in SA-CS_GEM NPs against A549 cells. IC50 for SA-CS_GEM NPs plunged with an increase in the time points from 24 to 72 h. Concentration-dependent haemolytic study confirmed significant haemocompatibility of SA-CS_GEM NPs. Pharmacokinetic study was performed on Sprague-Dawley rats and the kinetic parameters were calculated using PKSolver 2.0. Results demonstrated a consequential refinement of 2.98 times in modified SA-CS_GEM NPs with a significant increase in retention time, bioavailability and elimination half-life, and decrease in elimination rate constant and volume of distribution in comparison to CS_GEM NPs. Therefore, SA-CS shell core nanoparticles could be a beneficial approach to target and treat NSCLC (non-small cell lung cancer) and direct for research possibilities to target the other tumour cells.
Collapse
|
74
|
Marques P, Barry S, Carlsen E, Collier D, Ronaldson A, Grieve J, Dorward N, Mendoza N, Nair R, Muquit S, Grossman AB, Korbonits M. The expression of neural cell adhesion molecule and the microenvironment of pituitary neuroendocrine tumours. J Neuroendocrinol 2021; 33:e13052. [PMID: 34708902 DOI: 10.1111/jne.13052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022]
Abstract
The neural cell adhesion molecule (NCAM) has previously been studied in pituitary neuroendocrine tumours (PitNETs), but its role in tumour biology and aggressiveness remains controversial, and its relationship with the tumour microenvironment remains unknown. We aimed to characterise NCAM expression in PitNETs, to correlate this with clinico-pathological features, and to assess the role of various microenvironment components on NCAM expression. NCAM and immune cells were investigated by immunohistochemistry in 16 human non-functioning-PitNETs (NF-PitNETs) and eight somatotrophinomas, including macrophages (CD68, CD163, HLA-DR), cytotoxic (CD8) and T helper (CD4) lymphocytes, regulatory T cells (FOXP3), B cells (CD20), and neutrophils (neutrophil elastase). Five normal pituitaries were included for comparison. The cytokine secretome from these PitNETs and from PitNET-derived tumour-associated fibroblasts (TAFs) were assessed on culture supernatants using a multiplex immunoassay panel. There were no significant NCAM expression differences between PitNETs and normal pituitary, and no difference between types of pituitary tumours (NF-PitNETs vs. somatotrophinomas). There was no association between NCAM expression and different clinico-pathological features, including cavernous sinus invasion and Ki-67, nor with serum hormone levels. NCAM immunoreactivity correlated negatively with PitNET-derived CXCL10 (rho = -0.417; p = .042) and CX3CL1 (rho = -0.423; p = .040) levels. NCAM immunoreactivity was negatively correlated with TAF-derived fibroblast growth factor (FGF)-2 (rho = -0.632; p = .009), but not with other TAF-derived cytokines. Within the PitNET cohort, there were no correlations between NCAM immunoreactivity and immune infiltrates or ratios, although, within NF-PitNETs, NCAM expression was higher in tumours with more FOXP3+ cells. NCAM expression does not differ between PitNETs and normal pituitary, and does not appear to relate to tumour invasiveness or proliferation. However, our data suggest a possible role for cytokines in the modulation of NCAM expression in PitNETs, particularly CXCL10, CX3CL1 and FGF-2, but not for immune cell infiltrates.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Endocrinology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Sayka Barry
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - David Collier
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Amy Ronaldson
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Joan Grieve
- The National Hospital for Neurology and Neurosurgery, UCLH, NHS Trust, London, UK
| | - Neil Dorward
- The National Hospital for Neurology and Neurosurgery, UCLH, NHS Trust, London, UK
| | - Nigel Mendoza
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, UK
| | - Ramesh Nair
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, UK
| | - Samiul Muquit
- Department of Neurosurgery, Derriford Hospital, Plymouth, UK
| | - Ashley B Grossman
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
75
|
Jung J, Enterina JR, Bui DT, Mozaneh F, Lin PH, Nitin, Kuo CW, Rodrigues E, Bhattacherjee A, Raeisimakiani P, Daskhan GC, St. Laurent CD, Khoo KH, Mahal LK, Zandberg WF, Huang X, Klassen JS, Macauley MS. Carbohydrate Sulfation As a Mechanism for Fine-Tuning Siglec Ligands. ACS Chem Biol 2021; 16:2673-2689. [PMID: 34661385 DOI: 10.1021/acschembio.1c00501] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The immunomodulatory family of Siglecs recognizes sialic acid-containing glycans as "self", which is exploited in cancer for immune evasion. The biochemical nature of Siglec ligands remains incompletely understood, with emerging evidence suggesting the importance of carbohydrate sulfation. Here, we investigate how specific sulfate modifications affect Siglec ligands by overexpressing eight carbohydrate sulfotransferases (CHSTs) in five cell lines. Overexpression of three CHSTs─CHST1, CHST2, or CHST4─significantly enhance the binding of numerous Siglecs. Unexpectedly, two other CHSTs (Gal3ST2 and Gal3ST3) diminish Siglec binding, suggesting a new mode to modulate Siglec ligands via sulfation. Results are cell type dependent, indicating that the context in which sulfated glycans are presented is important. Moreover, a pharmacological blockade of N- and O-glycan maturation reveals a cell-type-specific pattern of importance for either class of glycan. Production of a highly homogeneous Siglec-3 (CD33) fragment enabled a mass-spectrometry-based binding assay to determine ≥8-fold and ≥2-fold enhanced affinity for Neu5Acα2-3(6-O-sulfo)Galβ1-4GlcNAc and Neu5Acα2-3Galβ1-4(6-O-sulfo)GlcNAc, respectively, over Neu5Acα2-3Galβ1-4GlcNAc. CD33 shows significant additivity in affinity (≥28-fold) for the disulfated ligand, Neu5Acα2-3(6-O-sulfo)Galβ1-4(6-O-sulfo)GlcNAc. Moreover, joint overexpression of CHST1 with CHST2 in cells greatly enhanced the binding of CD33 and several other Siglecs. Finally, we reveal that CHST1 is upregulated in numerous cancers, correlating with poorer survival rates and sodium chlorate sensitivity for the binding of Siglecs to cancer cell lines. These results provide new insights into carbohydrate sulfation as a general mechanism for tuning Siglec ligands on cells, including in cancer.
Collapse
Affiliation(s)
- Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Jhon R. Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G 2J7, Canada
| | - Duong T. Bui
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Fahima Mozaneh
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Po-Han Lin
- Departments of Chemistry and Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Nitin
- Department of Chemistry, The University of British Columbia, Kelowna, V1V 1V7, Canada
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Emily Rodrigues
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | | | | | - Gour C. Daskhan
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | | | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Lara K. Mahal
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Wesley F. Zandberg
- Department of Chemistry, The University of British Columbia, Kelowna, V1V 1V7, Canada
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - John S. Klassen
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
| | - Matthew S. Macauley
- Department of Chemistry, University of Alberta, Edmonton, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, T6G 2J7, Canada
| |
Collapse
|
76
|
Shi H, Chen Y, Li Y, Chen L, Wang H, Yang C, Ding L, Ju H. Hierarchical Fluorescence Imaging Strategy for Assessment of the Sialylation Level of Lipid Rafts on the Cell Membrane. Anal Chem 2021; 93:14643-14650. [PMID: 34698497 DOI: 10.1021/acs.analchem.1c02738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glycosylation is one of the most ubiquitous and complicated modifications of proteins and lipids. The revelation of glycosylation-mediated regulation mechanisms of biological processes relies critically on the tools that can reflect the spatial heterogeneity of cell surface glycans, for example, distinguishing glycans exhibited in lipid raft or nonraft domains. To achieve simultaneous visualization of raft and raft-harbored glycans on the cell surface, we combine specific raft recognition, glycan chemoselective labeling, and DNA dynamic hybridization techniques to develop a hierarchical fluorescence imaging strategy using N-acetyl-neuraminic acid (Sia) as the model sugar. We fabricate a raft probe and Sia probe for rafts and Sia, respectively. After specifically anchoring the two probes on the cell surface, the raft probe can be cyclically utilized to turn on the fluorescence of the Sia probe, only residing in rafts, via a proximity cascade DNA reaction. The duplex imaging capability for spatially relevant levels of biological structures enables the revelation of the reason for raft-confined Sia variation in different biological processes. Thus, this work provides an elegant and powerful tool for interrogation of the glycan regulation mechanisms on raft composition, organization, and functions and also contributes to the development of raft-carried glycoconjugate-based theranostic techniques.
Collapse
Affiliation(s)
- Huifang Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Younan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Chen Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| |
Collapse
|
77
|
Dobie C, Montgomery AP, Szabo R, Yu H, Skropeta D. Synthesis and biological evaluation of selective phosphonate-bearing 1,2,3-triazole-linked sialyltransferase inhibitors. RSC Med Chem 2021; 12:1680-1689. [PMID: 34778769 DOI: 10.1039/d1md00079a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/19/2021] [Indexed: 01/01/2023] Open
Abstract
The critical role of sialyltransferase (ST) enzymes in tumour cell growth and metastasis, as well as links to multi-drug and radiation resistance, has seen STs emerge as a target for potential antimetastatic cancer treatments. One promising class of ST inhibitors that improve upon the pharmacokinetic issues of previous inhibitors is the 1,2,3-triazole-linked transition-state analogues. Herein, we present the design and synthesis of a new generation of 1,2,3-triazole-linked sialyltransferase inhibitors, along with their biological evaluation demonstrating increased potency for phosphonate bearing compounds. The six most promising inhibitors presented in this work exhibited a greater number of binding modes for hST6Gal I over hST3Gal I, with K i ranging from 3-55 μM. This work highlights phosphonate bearing triazole-linked compounds as a promising class of synthetically accessible ST inhibitors that warrant further investigation.
Collapse
Affiliation(s)
- Christopher Dobie
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Andrew P Montgomery
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Rémi Szabo
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia
| | - Haibo Yu
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia .,Illawarra Health and Medical Research Institute Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong NSW 2522 Australia .,Illawarra Health and Medical Research Institute Wollongong NSW 2522 Australia
| |
Collapse
|
78
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
79
|
Tampa M, Nicolae I, Mitran CI, Mitran MI, Ene C, Matei C, Georgescu SR, Ene CD. Serum Sialylation Changes in Actinic Keratosis and Cutaneous Squamous Cell Carcinoma Patients. J Pers Med 2021; 11:jpm11101027. [PMID: 34683168 PMCID: PMC8538811 DOI: 10.3390/jpm11101027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 01/16/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC), a malignant proliferation of the cutaneous epithelium, is the second most common skin cancer after basal cell carcinoma (BCC). Unlike BCC, cSCC exhibits a greater aggressiveness and the ability to metastasize to any organ in the body. Chronic inflammation and immunosuppression are important processes linked to the development of cSCC. The tumor can occur de novo or from the histological transformation of preexisting actinic keratoses (AK). Malignant cells exhibit a higher amount of sialic acid in their membranes than normal cells, and changes in the amount, type, or linkage of sialic acid in malignant cell glycoconjugates are related to tumor progression and metastasis. The aim of our study was to investigate the sialyation in patients with cSCC and patients with AK. We have determined the serum levels of total sialic acid (TSA), lipid-bound sialic acid (LSA), beta-galactoside 2,6-sialyltransferase I (ST6GalI), and neuraminidase 3 (NEU3) in 40 patients with cSCC, 28 patients with AK, and 40 healthy subjects. Data analysis indicated a significant increase in serum levels of TSA (p < 0.001), LSA (p < 0.001), ST6GalI (p < 0.001), and NEU3 (p < 0.001) in the cSCC group compared to the control group, whereas in patients with AK only the serum level of TSA was significantly higher compared to the control group (p < 0.001). When the cSCC and AK groups were compared, significant differences between the serum levels of TSA (p < 0.001), LSA (p < 0.001), ST6GalI (p < 0.001) and NEU3 (p < 0.001) were found. The rate of synthesis of sialoglycoconjugates and their rate of enzymatic degradation, expressed by the ST6GalI/NEU3 ratio, is 1.64 times lower in the cSCC group compared to the control group (p < 0.01) and 1.53 times lower compared to the AK group (p < 0.01). The tumor diameter, depth of invasion, and Ki67 were associated with higher levels of TSA and LSA. These results indicate an aberrant sialylation in cSCC that correlates with tumor aggressiveness.
Collapse
Affiliation(s)
- Mircea Tampa
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.)
- Department of Dermatology, ‘Victor Babes’ Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| | - Ilinca Nicolae
- Department of Dermatology, ‘Victor Babes’ Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
- Correspondence: (I.N.); (S.R.G.)
| | - Cristina Iulia Mitran
- Department of Microbiology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.M.); (M.I.M.)
| | - Madalina Irina Mitran
- Department of Microbiology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.M.); (M.I.M.)
| | - Cosmin Ene
- Departments of Urology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Clara Matei
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.)
| | - Simona Roxana Georgescu
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.)
- Department of Dermatology, ‘Victor Babes’ Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
- Correspondence: (I.N.); (S.R.G.)
| | - Corina Daniela Ene
- Department of Nephrology, ‘Carol Davila’ Nephrology Hospital, 010731 Bucharest, Romania;
- Departments of Nephrology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
80
|
Abstract
The surface of every eukaryotic cell is coated in a dense layer of structurally diverse glycans that together comprise the glycocalyx, a key interface between intracellular biochemistry and the external environment. Many of the glycans within the glycocalyx terminate in anionic monosaccharides belonging to the sialic acid family. Advances in our understanding of the biological processes mediated by sialic acids at the interfaces between cells have catalyzed interest in metabolic, enzymatic, and chemical strategies to edit the total complement of cellular sialic acids-the sialome. Here, we review strategies for altering the composition of the sialome with particular focus on glycan structures and state-of-the-art tools.
Collapse
Affiliation(s)
- Landon J. Edgar
- Department of Pharmacology and Toxicology, The University of Toronto, Toronto, Ontario, Canada, M5S 1A8
| |
Collapse
|
81
|
Payazdan M, Khatami S, Galehdari H, Delfan N, Shafiei M, Heydaran S. The anti-inflammatory effects of sialic acid on the human glia cells by the upregulation of IL-4 and IL-10 genes' expressions. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
82
|
The T/Tn-Specific Helix pomatia Lectin Induces Cell Death in Lymphoma Cells Negative for T/Tn Antigens. Cancers (Basel) 2021; 13:cancers13174356. [PMID: 34503166 PMCID: PMC8431231 DOI: 10.3390/cancers13174356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Changes in glycosylation, such as incomplete synthesis and higher density of O-glycans on the cell surface, are frequently observed in cancer cells. Several types of truncated O-glycan structures, e.g., T/Tn antigens, are suspected to disrupt molecular interactions between tumor microenvironment and immune cells, for instance, facilitating cancer immune-escape. Therefore, numerous exogenous lectins targeting aberrant O-glycans are interesting tools for cancer diagnosis, prognosis, and therapy. However, the ability of exolectins to detect subtle alterations in the glycome of tumor cells and to interfere in tumor/healthy cell interactions remains largely unknown. The present article reports for the first time that the Helix pomatia (HPA) lectin, a well-known T/Tn-specific lectin, currently used as a tool in cancer diagnostics, kills Tn-positive leukemia cells and Tn-negative lymphoma cells but does not affect healthy lymphocytes. Thus, HPA could be used to discriminate between tumor and healthy cells, and detect subtle alterations in the glycosylation profile. Abstract Morniga G is a T/Tn-specific lectin, inducing cell death in Tn-positive leukemias but not in healthy lymphocytes. Helix pomatia lectin (HPA) is another T/Tn-specific lectin, currently used as tool for cancer diagnostics. The HPA-mediated tumor cell death was evaluated on human leukemia and mouse lymphoma cells, and compared to the effect of Morniga G. Both lectins induced an equivalent percentage of cell death in Tn-positive Jurkat human leukemia. In contrast, EL4 mouse lymphoma resisted Morniga G-mediated cytotoxicity but were killed by HPA at concentrations of 2.5 μg/mL (0.032 nM) and higher. In both malignant cells, HPA-mediated cell death showed features compatible with apoptosis (annexin-externalization, caspase-activation, mitochondrial membrane depolarization, and ROS production). Cytometry analysis indicated that EL4 cells are T/Tn-negative. Because previous results showed a high amount of N-acetylgalactosamine (GalNAc, sugar present in Tn antigen) on EL4 cell surface, this GalNAc could be involved in the formation of truncated O-glycans other than the T/Tn residues. When compared to Morniga G, bioinformatic analysis suggested that HPA benefits from an extended carbohydrate-binding site, better adapted than Morniga G to the accommodation of more complex branched and truncated O-glycans (such as core 2). Finally, HPA killed EL4 cells but not healthy lymphocytes in a mixture of lymphoma cells + lymphocytes, suggesting that HPA selectively triggers tumor cell death.
Collapse
|
83
|
Xu Z, Zhang Y, Ocansey DKW, Wang B, Mao F. Glycosylation in Cervical Cancer: New Insights and Clinical Implications. Front Oncol 2021; 11:706862. [PMID: 34485140 PMCID: PMC8415776 DOI: 10.3389/fonc.2021.706862] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Cervical cancer has become the most frequent female malignancy and presents as a general health challenge in many countries undergoing economic development. Various human papillomaviruses (HPV) types have appeared as one of the most critically identifiable causes of widespread cervical cancers. Conventional cervical cytological inspection has limitations of variable sensitivity according to cervical cytology. Glycobiology has been fundamental in related exploration in various gynecologic and reproductive fields and has contributed to our understanding of cervical cancer. It is associated with altered expression of N-linked glycan as well as abnormal expression of terminal glycan structures. The analytical approaches available to determine serum and tissue glycosylation, as well as potential underlying molecular mechanisms involved in the cellular glycosylation alterations, are monitored. Moreover, cellular glycosylation influences various aspects of cervical cancer biology, ranging from cell surface expressions, cell-cell adhesion, cancer signaling, cancer diagnosis, and management. In general, discoveries in glycan profiling make it technically reproducible and affordable to perform serum glycoproteomic analyses and build on previous work exploring an expanded variety of glycosylation markers in the majority of cervical cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
84
|
Li D, Mukhopadhyay S. A three-pocket model for substrate coordination and selectivity by the nucleotide sugar transporters SLC35A1 and SLC35A2. J Biol Chem 2021; 297:101069. [PMID: 34384782 PMCID: PMC8411240 DOI: 10.1016/j.jbc.2021.101069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 10/25/2022] Open
Abstract
The CMP-sialic acid transporter SLC35A1 and UDP-galactose transporter SLC35A2 are two well-characterized nucleotide sugar transporters with distinctive substrate specificities. Mutations in either induce congenital disorders of glycosylation. Despite the biomedical relevance, mechanisms of substrate specificity are unclear. To address this critical issue, we utilized a structure-guided mutagenesis strategy and assayed a series of SLC35A2 and SLC35A1 mutants using a rescue approach. Our results suggest that three pockets in the central cavity of each transporter provide substrate specificity. The pockets comprise (1) nucleobase (residues E52, K55, and Y214 of SLC35A1; E75, K78, N235, and G239 of SLC35A2); (2) middle (residues Q101, N102, and T260 of SLC35A1; Q125, N126, Q129, Y130, and Q278 of SLC35A2); and (3) sugar (residues K124, T128, S188, and K272 of SLC35A1; K148, T152, S213, and K297 of SLC35A2) pockets. Within these pockets, two components appear to be especially critical for substrate specificity. Y214 (for SLC35A1) and G239 (for SLC35A2) in the nucleobase pocket appear to discriminate cytosine from uracil. Furthermore, Q129 and Q278 of SLC35A2 in the middle pocket appear to interact specifically with the β-phosphate of UDP while the corresponding A105 and A253 residues in SLC35A1 do not interact with CMP, which lacks a β-phosphate. Overall, our findings contribute to a molecular understanding of substrate specificity and coordination in SLC35A1 and SLC35A2, and have important implications for the understanding and treatment of diseases associated with mutations or dysregulations of these two transporters.
Collapse
Affiliation(s)
- Danyang Li
- Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, Institute for Cellular & Molecular Biology, and Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712.
| |
Collapse
|
85
|
Friedman DJ, Crotts SB, Shapiro MJ, Rajcula M, McCue S, Liu X, Khazaie K, Dong H, Shapiro VS. ST8Sia6 Promotes Tumor Growth in Mice by Inhibiting Immune Responses. Cancer Immunol Res 2021; 9:952-966. [PMID: 34074677 PMCID: PMC8338779 DOI: 10.1158/2326-6066.cir-20-0834] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/05/2021] [Accepted: 05/26/2021] [Indexed: 01/27/2023]
Abstract
Many tumors exhibit increased incorporation of sialic acids into cell-surface glycans, which impact the tumor microenvironment. Sialic acid immunoglobulin-like lectins (Siglec) are receptors that recognize sialic acids and modulate immune responses, including responses to tumors. However, the roles of individual sialyltransferases in tumorigenesis and tumor growth are not well understood. Here, we examined the sialyltransferase ST8Sia6, which generated α2,8-linked disialic acids that bind to murine Siglec-E and human Siglec-7 and -9. Increased ST8Sia6 expression was found on many human tumors and associated with decreased survival in several cancers, including colon cancer. Because of this, we engineered MC38 and B16-F10 tumor lines to express ST8Sia6. ST8Sia6-expressing MC38 and B16-F10 tumors exhibited faster growth and led to decreased survival, which required host Siglec-E. ST8Sia6 expression on tumors also altered macrophage polarization toward M2, including upregulation of the immune modulator arginase, which also required Siglec-E. ST8Sia6 also accelerated tumorigenesis in a genetically engineered, spontaneous murine model of colon cancer, decreasing survival from approximately 6 months to 67 days. Thus, ST8Sia6 expression on tumors inhibits antitumor immune responses to accelerate tumor growth.
Collapse
Affiliation(s)
| | | | | | | | - Shaylene McCue
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Xin Liu
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | | | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
- Department of Urology, College of Medicine, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
86
|
Seo N, Ko J, Lee D, Jeong H, Oh MJ, Kim U, Lee DH, Kim J, Choi YJ, An HJ. In-depth characterization of non-human sialic acid (Neu5Gc) in human serum using label-free ZIC-HILIC/MRM-MS. Anal Bioanal Chem 2021; 413:5227-5237. [PMID: 34235565 DOI: 10.1007/s00216-021-03495-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023]
Abstract
Sialic acid Neu5Gc, a non-human glycan, is recognized as a new harmful substance that can cause vascular disease and cancer. Humans are unable to synthesize Neu5Gc due to a genetic defect that converts Neu5Ac to Neu5Gc, but Neu5Gc is often observed in human biological samples. Therefore, the demand for accurately measuring the amount of Neu5Gc present in human blood or tissues is rapidly increasing, but there is still no method to reliably quantify trace amounts of a non-human sugar. In particular, selective isolation and detection of Neu5Gc from human serum is analytically challenging due to the presence of excess sialic acid Neu5Ac, which has physicochemical properties very similar to Neu5Gc. Herein, we developed the label-free approach based on ZIC-HILIC/MRM-MS that can enrich sialic acids released from human serum and simultaneously monitor Neu5Ac and Neu5Gc. The combination of complete separation of Neu5Gc from abundant Neu5Ac by hydrophilic and electrostatic interactions with selective monitoring of structure-specific cross-ring cleavage ions generated by negative CID-MS/MS was remarkably effective for quantification of Neu5Ac and Neu5Gc at the femtomole level. Indeed, we were able to successfully determine the absolute quantitation of Neu5Gc from 30 healthy donors in the range of 3.336 ± 1.252 pg/μL (mean ± SD), 10,000 times lower than Neu5Ac. In particular, analysis of sialic acids in protein-free serum revealed that both Neu5Ac and Neu5G are mostly bound to proteins and/or lipids, but not in free form. In addition, the correlation between expression level of Neu5Gc and biological factors such as BMI, age, and sex was investigated. This method can be widely used in studies requiring sialic acid-related measurements such as disease diagnosis or prediction of immunogenicity in biopharmaceuticals as it is both fast and highly sensitive.
Collapse
Affiliation(s)
- Nari Seo
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea
| | - Jaekyoung Ko
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea
| | - Daum Lee
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea
| | - Heejin Jeong
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea
| | - Myung Jin Oh
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea
| | - Unyong Kim
- Biocomplete Co., Ltd., Seoul, 08389, Republic of Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Budang Hospital, Seongnam, 13620, Republic of Korea
| | - Jaehan Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Yoon Jin Choi
- Department of Internal Medicine, Seoul National University Budang Hospital, Seongnam, 13620, Republic of Korea. .,Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea. .,Asia Glycomics Reference Site, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
87
|
Krick S, Helton ES, Easter M, Bollenbecker S, Denson R, Zaharias R, Cochran P, Vang S, Harris E, Wells JM, Barnes JW. ST6GAL1 and α2-6 Sialylation Regulates IL-6 Expression and Secretion in Chronic Obstructive Pulmonary Disease. Front Immunol 2021; 12:693149. [PMID: 34290711 PMCID: PMC8287524 DOI: 10.3389/fimmu.2021.693149] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/21/2021] [Indexed: 01/23/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a systemic disease strongly associated with cigarette smoking, airway inflammation, and acute disease exacerbations. Changes in terminal sialylation and fucosylation of asparagine (N)-linked glycans have been documented in COPD, but the role that glycosyltransferases may play in the regulation of N-linked glycans in COPD has not been fully elucidated. Recent studies suggest that modulation of ST6GAL1 (ST6 beta-galactoside alpha-2,6-sialyltransferase-1), which catalyzes terminal α2-6 sialylation of cellular proteins, may regulate inflammation and contribute to COPD phenotype(s). Interestingly, it has been previously demonstrated that ST6GAL1, a Golgi resident protein, can be proteolytically processed by BACE1 (beta-site amyloid precursor protein cleaving enzyme-1) to a circulating form that retains activity. In this study, we showed that loss of ST6GAL1 expression increased interleukin (IL)-6 expression and secretion in human bronchial epithelial cells (HBECs). Furthermore, exposure to cigarette smoke medium/extract (CSE) or BACE1 inhibition resulted in decreased ST6GAL1 secretion, reduced α2-6 sialylation, and increased IL-6 production in HBECs. Analysis of plasma ST6GAL1 levels in a small COPD patient cohort demonstrated an inverse association with prospective acute exacerbations of COPD (AECOPD), while IL-6 was positively associated. Altogether, these results suggest that reduced ST6GAL1 and α2-6 sialylation augments IL-6 expression/secretion in HBECs and is associated with poor clinical outcomes in COPD.
Collapse
Affiliation(s)
- Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - E. Scott Helton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rebecca Denson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rennan Zaharias
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Phillip Cochran
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shia Vang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Elex Harris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - James M. Wells
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Lung Health Center, Birmingham, AL, United States
- Birmingham VA Medical Center, Birmingham, AL, United States
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
88
|
Brücher D, Franc V, Smith SN, Heck AJR, Plückthun A. Malignant tissues produce divergent antibody glycosylation of relevance for cancer gene therapy effectiveness. MAbs 2021; 12:1792084. [PMID: 32643525 PMCID: PMC7531505 DOI: 10.1080/19420862.2020.1792084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gene therapy approaches now allow for the production of therapeutic antibodies by healthy or cancerous human tissues directly in vivo, and, with an increasing number of gene delivery methods available, the cell type for expression can be chosen. Yet, little is known about the biophysical changes introduced by expressing antibodies from producer cells or tissues targeted by gene therapy approaches, nor about the consequences for the type of glycosylation. The effects of different glycosylation on therapeutic antibodies have been well studied by controlling their glycan compositions in non-human mammalian production cells, i.e., Chinese hamster ovary cells. Therefore, we investigated the glycosylation state of clinically approved antibodies secreted from cancer tissues frequently targeted by in vivo gene therapy, using native mass spectrometry and glycoproteomics. We found that antibody sialylation and fucosylation depended on the producer tissue and the antibody isotype, allowing us to identify optimal producer cell types according to the desired mode of action of the antibody. Furthermore, we discovered that high amounts (>20%) of non-glycosylated antibodies were produced in cells sensitive to the action of the produced antibodies. Different glycosylation in different producer cells can translate into an altered potency of in-vivo produced antibodies, depending on the desired mode of action, and can affect their serum half-lives. These results increase our knowledge about antibodies produced from cells targeted by gene therapy, enabling development of improved cancer gene therapy vectors that can include in vivo glycoengineering of expressed antibodies to optimize their efficacies, depending on the desired mode of action.
Collapse
Affiliation(s)
- Dominik Brücher
- Department of Biochemistry, University of Zurich , Zurich, Switzerland
| | - Vojtech Franc
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht , Utrecht, The Netherlands.,Netherlands Proteomics Center , Utrecht, The Netherlands
| | - Sheena N Smith
- Department of Biochemistry, University of Zurich , Zurich, Switzerland
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht , Utrecht, The Netherlands.,Netherlands Proteomics Center , Utrecht, The Netherlands
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich , Zurich, Switzerland
| |
Collapse
|
89
|
Lakshmanan I, Chaudhary S, Vengoji R, Seshacharyulu P, Rachagani S, Carmicheal J, Jahan R, Atri P, Chirravuri‐Venkata R, Gupta R, Marimuthu S, Perumal N, Rauth S, Kaur S, Mallya K, Smith LM, Lele SM, Ponnusamy MP, Nasser MW, Salgia R, Batra SK, Ganti AK. ST6GalNAc-I promotes lung cancer metastasis by altering MUC5AC sialylation. Mol Oncol 2021; 15:1866-1881. [PMID: 33792183 PMCID: PMC8253099 DOI: 10.1002/1878-0261.12956] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/19/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related mortality. However, the molecular mechanisms associated with the development of metastasis are poorly understood. Understanding the biology of LC metastasis is critical to unveil the molecular mechanisms for designing targeted therapies. We developed two genetically engineered LC mouse models KrasG12D/+ ; Trp53R172H/+ ; Ad-Cre (KPA) and KrasG12D/+ ; Ad-Cre (KA). Survival analysis showed significantly (P = 0.0049) shorter survival in KPA tumor-bearing mice as compared to KA, suggesting the aggressiveness of the model. Our transcriptomic data showed high expression of N-acetylgalactosaminide alpha-2, 6-sialyltransferase 1 (St6galnac-I) in KPA compared to KA tumors. ST6GalNAc-I is an O-glycosyltransferase, which catalyzes the addition of sialic acid to the initiating GalNAc residues forming sialyl Tn (STn) on glycoproteins, such as mucins. Ectopic expression of species-specific p53 mutants in the syngeneic mouse and human LC cells led to increased cell migration and high expression of ST6GalNAc-I, STn, and MUC5AC. Immunoprecipitation of MUC5AC in the ectopically expressing p53R175H cells exhibited higher affinity toward STn. In addition, ST6GalNAc-I knockout (KO) cells also showed decreased migration, possibly due to reduced glycosylation of MUC5AC as observed by low STn on the glycoprotein. Interestingly, ST6GalNAc-I KO cells injected mice developed less liver metastasis (P = 0.01) compared to controls, while colocalization of MUC5AC and STn was observed in the liver metastatic tissues of control mice. Collectively, our findings support the hypothesis that mutant p53R175H mediates ST6GalNAc-I expression, leading to the sialyation of MUC5AC, and thus contribute to LC liver metastasis.
Collapse
Affiliation(s)
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | | | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Rahat Jahan
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Pranita Atri
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | | | - Rohitesh Gupta
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Saravanakumar Marimuthu
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Naveenkumar Perumal
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Kavita Mallya
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Lynette M. Smith
- Department of BiostatisticsCollege of Public HealthUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Subodh M. Lele
- Department of Pathology and MicrobiologyUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Moorthy P. Ponnusamy
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
- Eppley Institute for Research in Cancer and Allied DiseasesOmahaNEUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Mohd W. Nasser
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics ResearchCity of Hope Comprehensive Cancer CenterBeckman Research InstituteDuarteCAUSA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
- Eppley Institute for Research in Cancer and Allied DiseasesOmahaNEUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNEUSA
| | - Apar Kishor Ganti
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNEUSA
- Fred & Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNEUSA
- Department of Internal MedicineVA Nebraska Western Iowa Health Care SystemUniversity of Nebraska Medical CenterOmahaNEUSA
| |
Collapse
|
90
|
Cockram TOJ, Dundee JM, Popescu AS, Brown GC. The Phagocytic Code Regulating Phagocytosis of Mammalian Cells. Front Immunol 2021; 12:629979. [PMID: 34177884 PMCID: PMC8220072 DOI: 10.3389/fimmu.2021.629979] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/18/2021] [Indexed: 01/21/2023] Open
Abstract
Mammalian phagocytes can phagocytose (i.e. eat) other mammalian cells in the body if they display certain signals, and this phagocytosis plays fundamental roles in development, cell turnover, tissue homeostasis and disease prevention. To phagocytose the correct cells, phagocytes must discriminate which cells to eat using a 'phagocytic code' - a set of over 50 known phagocytic signals determining whether a cell is eaten or not - comprising find-me signals, eat-me signals, don't-eat-me signals and opsonins. Most opsonins require binding to eat-me signals - for example, the opsonins galectin-3, calreticulin and C1q bind asialoglycan eat-me signals on target cells - to induce phagocytosis. Some proteins act as 'self-opsonins', while others are 'negative opsonins' or 'phagocyte suppressants', inhibiting phagocytosis. We review known phagocytic signals here, both established and novel, and how they integrate to regulate phagocytosis of several mammalian targets - including excess cells in development, senescent and aged cells, infected cells, cancer cells, dead or dying cells, cell debris and neuronal synapses. Understanding the phagocytic code, and how it goes wrong, may enable novel therapies for multiple pathologies with too much or too little phagocytosis, such as: infectious disease, cancer, neurodegeneration, psychiatric disease, cardiovascular disease, ageing and auto-immune disease.
Collapse
Affiliation(s)
| | | | | | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
91
|
Hussein NA, Malla S, Pasternak MA, Terrero D, Brown NG, Ashby CR, Assaraf YG, Chen ZS, Tiwari AK. The role of endolysosomal trafficking in anticancer drug resistance. Drug Resist Updat 2021; 57:100769. [PMID: 34217999 DOI: 10.1016/j.drup.2021.100769] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/10/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
Multidrug resistance (MDR) remains a major obstacle towards curative treatment of cancer. Despite considerable progress in delineating the basis of intrinsic and acquired MDR, the underlying molecular mechanisms remain to be elucidated. Emerging evidences suggest that dysregulation in endolysosomal compartments is involved in mediating MDR through multiple mechanisms, such as alterations in endosomes, lysosomes and autophagosomes, that traffic and biodegrade the molecular cargo through macropinocytosis, autophagy and endocytosis. For example, altered lysosomal pH, in combination with transcription factor EB (TFEB)-mediated lysosomal biogenesis, increases the sequestration of hydrophobic anti-cancer drugs that are weak bases, thereby producing an insufficient and off-target accumulation of anti-cancer drugs in MDR cancer cells. Thus, the use of well-tolerated, alkalinizing compounds that selectively block Vacuolar H⁺-ATPase (V-ATPase) may be an important strategy to overcome MDR in cancer cells and increase chemotherapeutic efficacy. Other mechanisms of endolysosomal-mediated drug resistance include increases in the expression of lysosomal proteases and cathepsins that are involved in mediating carcinogenesis and chemoresistance. Therefore, blocking the trafficking and maturation of lysosomal proteases or direct inhibition of cathepsin activity in the cytosol may represent novel therapeutic modalities to overcome MDR. Furthermore, endolysosomal compartments involved in catabolic pathways, such as macropinocytosis and autophagy, are also shown to be involved in the development of MDR. Here, we review the role of endolysosomal trafficking in MDR development and discuss how targeting endolysosomal pathways could emerge as a new therapeutic strategy to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Noor A Hussein
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Mariah A Pasternak
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Noah G Brown
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY, USA.
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, 43614, OH, USA; Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, 43614, OH, USA.
| |
Collapse
|
92
|
Mass Spectrometry-Based Glycoproteomics and Prostate Cancer. Int J Mol Sci 2021; 22:ijms22105222. [PMID: 34069262 PMCID: PMC8156230 DOI: 10.3390/ijms22105222] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
Aberrant glycosylation has long been known to be associated with cancer, since it is involved in key mechanisms such as tumour onset, development and progression. This review will focus on protein glycosylation studies in cells, tissue, urine and serum in the context of prostate cancer. A dedicated section will cover the glycoforms of prostate specific antigen, the molecule that, despite some important limitations, is routinely tested for helping prostate cancer diagnosis. Our aim is to provide readers with an overview of mass spectrometry-based glycoproteomics of prostate cancer. From this perspective, the first part of this review will illustrate the main strategies for glycopeptide enrichment and mass spectrometric analysis. The molecular information obtained by glycoproteomic analysis performed by mass spectrometry has led to new insights into the mechanism linking aberrant glycosylation to cancer cell proliferation, migration and immunoescape.
Collapse
|
93
|
Guo X, Elkashef SM, Patel A, Ribeiro Morais G, Shnyder SD, Loadman PM, Patterson LH, Falconer RA. An assay for quantitative analysis of polysialic acid expression in cancer cells. Carbohydr Polym 2021; 259:117741. [PMID: 33674001 DOI: 10.1016/j.carbpol.2021.117741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 10/22/2022]
Abstract
Polysialic acid (polySia) is a linear polysaccharide comprised of N-acetylneuraminic acid residues and its over-expression in cancer cells has been correlated with poor clinical prognosis. An assay has been developed for quantitative analysis of cellular polySia expression. This was achieved by extracting and purifying released polySia from glycoproteins by mild acid hydrolysis and optimised organic extraction. The polySia was further hydrolysed into Sia monomers, followed by fluorescent labelling and quantitative analysis. The assay was qualified utilising endoneuraminidase-NF to remove polySia from the surface of C6-ST8SiaII cancer cells (EC50 = 2.13 ng/mL). The result was comparable to that obtained in a polySia-specific cellular ELISA assay. Furthermore, the assay proved suitable for evaluation of changes in polySia expression following treatment with a small molecule inhibitor of polysialylation. Given the importance of polySia in multiple disease states, notably cancer, this is a potentially vital tool with applications in the fields of drug discovery and glycobiology.
Collapse
Affiliation(s)
- Xiaoxiao Guo
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Sara M Elkashef
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Anjana Patel
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Goreti Ribeiro Morais
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Steven D Shnyder
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Paul M Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Laurence H Patterson
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Robert A Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom.
| |
Collapse
|
94
|
Li RJE, de Haas A, Rodríguez E, Kalay H, Zaal A, Jimenez CR, Piersma SR, Pham TV, Henneman AA, de Goeij-de Haas RR, van Vliet SJ, van Kooyk Y. Quantitative Phosphoproteomic Analysis Reveals Dendritic Cell- Specific STAT Signaling After α2-3-Linked Sialic Acid Ligand Binding. Front Immunol 2021; 12:673454. [PMID: 33968084 PMCID: PMC8100677 DOI: 10.3389/fimmu.2021.673454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Dendritic cells (DCs) are key initiators of the adaptive immunity, and upon recognition of pathogens are able to skew T cell differentiation to elicit appropriate responses. DCs possess this extraordinary capacity to discern external signals using receptors that recognize pathogen-associated molecular patterns. These can be glycan-binding receptors that recognize carbohydrate structures on pathogens or pathogen-associated patterns that additionally bind receptors, such as Toll-like receptors (TLRs). This study explores the early signaling events in DCs upon binding of α2-3 sialic acid (α2-3sia) that are recognized by Immune inhibitory Sialic acid binding immunoglobulin type lectins. α2-3sias are commonly found on bacteria, e.g. Group B Streptococcus, but can also be expressed by tumor cells. We investigated whether α2-3sia conjugated to a dendrimeric core alters DC signaling properties. Through phosphoproteomic analysis, we found differential signaling profiles in DCs after α2-3sia binding alone or in combination with LPS/TLR4 co-stimulation. α2-3sia was able to modulate the TLR4 signaling cascade, resulting in 109 altered phosphoproteins. These phosphoproteins were annotated to seven biological processes, including the regulation of the IL-12 cytokine pathway. Secretion of IL-10, the inhibitory regulator of IL-12 production, by DCs was found upregulated after overnight stimulation with the α2-3sia dendrimer. Analysis of kinase activity revealed altered signatures in the JAK-STAT signaling pathway. PhosphoSTAT3 (Ser727) and phosphoSTAT5A (Ser780), involved in the regulation of the IL-12 pathway, were both downregulated. Flow cytometric quantification indeed revealed de- phosphorylation over time upon stimulation with α2-3sia, but no α2-6sia. Inhibition of both STAT3 and -5A in moDCs resulted in a similar cytokine secretion profile as α-3sia triggered DCs. Conclusively, this study revealed a specific alteration of the JAK-STAT pathway in DCs upon simultaneous α2-3sia and LPS stimulation, altering the IL10:IL-12 cytokine secretion profile associated with reduction of inflammation. Targeted control of the STAT phosphorylation status is therefore an interesting lead for the abrogation of immune escape that bacteria or tumors impose on the host.
Collapse
Affiliation(s)
- Rui-Jún Eveline Li
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Aram de Haas
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ernesto Rodríguez
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Anouk Zaal
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sander R Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Thang V Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Alex A Henneman
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Richard R de Goeij-de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
95
|
Rawal P, Zhao L. Sialometabolism in Brain Health and Alzheimer's Disease. Front Neurosci 2021; 15:648617. [PMID: 33867926 PMCID: PMC8044809 DOI: 10.3389/fnins.2021.648617] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acids refer to a unique family of acidic sugars with a 9-carbon backbone that are mostly found as terminal residues in glycan structures of glycoconjugates including both glycoproteins and glycolipids. The highest levels of sialic acids are expressed in the brain where they regulate neuronal sprouting and plasticity, axon myelination and myelin stability, as well as remodeling of mature neuronal connections. Moreover, sialic acids are the sole ligands for microglial Siglecs (sialic acid-binding immunoglobulin-type lectins), and sialic acid-Siglec interactions have been indicated to play a critical role in the regulation of microglial homeostasis in a healthy brain. The recent discovery of CD33, a microglial Siglec, as a novel genetic risk factor for late-onset Alzheimer's disease (AD), highlights the potential role of sialic acids in the development of microglial dysfunction and neuroinflammation in AD. Apart from microglia, sialic acids have been found to be involved in several other major changes associated with AD. Elevated levels of serum sialic acids have been reported in AD patients. Alterations in ganglioside (major sialic acid carrier) metabolism have been demonstrated as an aggravating factor in the formation of amyloid pathology in AD. Polysialic acids are linear homopolymers of sialic acids and have been implicated to be an important regulator of neurogenesis that contributes to neuronal repair and recovery from neurodegeneration such as in AD. In summary, this article reviews current understanding of neural functions of sialic acids and alterations of sialometabolism in aging and AD brains. Furthermore, we discuss the possibility of looking at sialic acids as a promising novel therapeutic target for AD intervention.
Collapse
Affiliation(s)
- Punam Rawal
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
96
|
Yang D, Yang L, Cai J, Hu X, Li H, Zhang X, Zhang X, Chen X, Dong H, Nie H, Li Y. A sweet spot for macrophages: Focusing on polarization. Pharmacol Res 2021; 167:105576. [PMID: 33771700 DOI: 10.1016/j.phrs.2021.105576] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/21/2022]
Abstract
Macrophages are a type of functionally plastic cells that can create a pro-/anti-inflammatory microenvironment for organs by producing different kinds of cytokines, chemokines, and growth factors to regulate immunity and inflammatory responses. In addition, they can also be induced to adopt different phenotypes in response to extracellular and intracellular signals, a process defined as M1/M2 polarization. Growing evidence indicates that glycobiology is closely associated with this polarization process. In this research, we review studies of the roles of glycosylation, glucose metabolism, and key lectins in the regulation of macrophages function and polarization to provide a new perspective for immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110000, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaxin Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaohan Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xinghe Chen
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haiyang Dong
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
97
|
Suleiman RB, Muhammad A, Umara IA, Ibrahima MA, Erukainure OL, Forcados GE, Katsayal SB. Kolaviron Ameliorates 7, 12-Dimethylbenzanthracene - Induced Mammary Damage in Female Wistar Rats. Anticancer Agents Med Chem 2021; 22:181-192. [PMID: 34225638 DOI: 10.2174/1871520621666210322101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/27/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Kolaviron (KV) is a flavonoid rich portion obtained from Garcinia kola seeds with a number of reported pharmacological effects. However, its ameliorative effects on 7,12-Dimethylbenzanthracene (DMBA)-induced mammary damage has not been fully investigated, despite the reported use of the seeds in the treatment of inflammatory related disorders. OBJECTIVE To evaluate the ameliorative effects of KV on DMBA-induced mammary damage in female Wistar rats. METHODS Forty-nine (49) female Wistar rats were randomly assigned into seven groups of seven rats each. DMBA was administered orally to rats in five of the groups as a single dose of 80 mg/kg body wt while the remaining two groups received the vehicle. The rats were palpated weekly for 3 months to monitor tumor formation. After 3 months of DMBA administration, 1 ml of blood was collected to assay for estrogen receptor- α (ER-α) level. Thereafter, the vehicle (dimethyl sulfoxide) was daily administered to the negative control and positive control groups for the 14 days duration of the experiment while three groups were each given a daily oral dose of 50, 100 and 200 mg/kg body wt of KV for the duration of the experiment. The last DMBA-induced group received 10 mg/kg body wt of the standard drug tamoxifen twice in a week and the remaining DMBA-free group received 200 mg/kg body wt KV. Subsequently, the animals were humanly sacrificed and ER-α, sialic acids, sialidase, sialyltransferase levels were assay for in blood and mammary tissues followed by histopathological examinations. RESULTS Significantly higher levels of estrogen receptor-α (ER-α), formation of lobular neoplastic cells, epithelial hyperplasia, lymphocyte infiltration and increased sialylation were detected in DMBA-induced rats. Treatment with KV at 50, 100 and 200 mg/kg body weight resulted in a significant (p<0.05) decrease in ER-α level, significantly (p<0.05) lower free serum sialic acid (21.1%), total sialic acid level of the mammary tissue (21.57%), sialyltransferase activity (30.83%) as well as mRNA level of the sialyltransferase gene (ST3Gal1) were observed after KV interventions. CONCLUSION The findings suggest that KV could be further explored in targeting DMBA-induced mammary damage implicated in mammary carcinogenesis.
Collapse
Affiliation(s)
- Rabiatu B Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ismaila A Umara
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed A Ibrahima
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein 9300. South Africa
| | - Gilead E Forcados
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sanusi B Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| |
Collapse
|
98
|
Achilli S, Berthet N, Renaudet O. Antibody recruiting molecules (ARMs): synthetic immunotherapeutics to fight cancer. RSC Chem Biol 2021; 2:713-724. [PMID: 34212148 PMCID: PMC8190906 DOI: 10.1039/d1cb00007a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Antibody-recruiting molecules (ARMs) are one of the most promising tools to redirect the immune response towards cancer cells. In this review, we aim to highlight the recent advances in the field. We will illustrate the advantages of different ARM approaches and emphasize the importance of a multivalent presentation of the binding units. Antibody-recruiting molecules (ARMs) are one of the most promising tools to redirect the immune response towards cancer cells.![]()
Collapse
Affiliation(s)
- Silvia Achilli
- Univ. Grenoble Alpes, CNRS DCM UMR 5250 F-38000 Grenoble France
| | | | | |
Collapse
|
99
|
Rodriguez E, Boelaars K, Brown K, Eveline Li RJ, Kruijssen L, Bruijns SCM, van Ee T, Schetters STT, Crommentuijn MHW, van der Horst JC, van Grieken NCT, van Vliet SJ, Kazemier G, Giovannetti E, Garcia-Vallejo JJ, van Kooyk Y. Sialic acids in pancreatic cancer cells drive tumour-associated macrophage differentiation via the Siglec receptors Siglec-7 and Siglec-9. Nat Commun 2021; 12:1270. [PMID: 33627655 PMCID: PMC7904912 DOI: 10.1038/s41467-021-21550-4] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022] Open
Abstract
Changes in glycosylation during tumour progression are a key hallmark of cancer. One of the glycan moieties generally overexpressed in cancer are sialic acids, which can induce immunomodulatory properties via binding to Siglec receptors. We here show that Pancreatic Ductal Adenocarcinoma (PDAC) tumour cells present an increased sialylation that can be recognized by Siglec-7 and Siglec-9 on myeloid cells. We identified the expression of the α2,3 sialyltransferases ST3GAL1 and ST3GAL4 as main contributor to the synthesis of ligands for Siglec-7 and Siglec-9 in tumour cells. Analysing the myeloid composition in PDAC, using single cell and bulk transcriptomics data, we identified monocyte-derived macrophages as contributors to the poor clinical outcome. Tumour-derived sialic acids dictate monocyte to macrophage differentiation via signalling through Siglec-7 and Siglec-9. Moreover, triggering of Siglec-9 in macrophages reduce inflammatory programmes, while increasing PD-L1 and IL-10 expression, illustrating that sialic acids modulate different myeloid cells. This work highlights a critical role for sialylated glycans in controlling immune suppression and provides new potential targets for cancer immunotherapy in PDAC.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kelly Boelaars
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kari Brown
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - R J Eveline Li
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Laura Kruijssen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sven C M Bruijns
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Thomas van Ee
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sjoerd T T Schetters
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Matheus H W Crommentuijn
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Joost C van der Horst
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Nicole C T van Grieken
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, Netherlands
| | - Sandra J van Vliet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Geert Kazemier
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Juan J Garcia-Vallejo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.
| |
Collapse
|
100
|
Wielgat P, Wawrusiewicz-Kurylonek N, Czarnomysy R, Rogowski K, Bielawski K, Car H. The Paired Siglecs in Brain Tumours Therapy: The Immunomodulatory Effect of Dexamethasone and Temozolomide in Human Glioma In Vitro Model. Int J Mol Sci 2021; 22:ijms22041791. [PMID: 33670244 PMCID: PMC7916943 DOI: 10.3390/ijms22041791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The paired sialic acid-binding immunoglobulin like lectins (Siglecs) are characterized by similar cellular distribution and ligand recognition but opposing signalling functions attributed to different intracellular sequences. Since sialic acid—Siglec axis are known to control immune homeostasis, the imbalance between activatory and inhibitory mechanisms of glycan-dependent immune control is considered to promote pathology. The role of sialylation in cancer is described, however, its importance in immune regulation in gliomas is not fully understood. The experimental and clinical observation suggest that dexamethasone (Dex) and temozolomide (TMZ), used in the glioma management, alter the immunity within the tumour microenvironment. Using glioma-microglia/monocytes transwell co-cultures, we investigated modulatory action of Dex/TMZ on paired Siglecs. Based on real-time PCR and flow cytometry, we found changes in SIGLEC genes and their products. These effects were accompanied by altered cytokine profile and immune cells phenotype switching measured by arginases expression. Additionally, the exposure to Dex or TMZ increased the binding of inhibitory Siglec-5 and Siglec-11 fusion proteins to glioma cells. Our study suggests that the therapy-induced modulation of the interplay between sialoglycans and paired Siglecs, dependently on patient’s phenotype, is of particular signification in the immune surveillance in the glioma management and may be useful in glioma patient’s therapy plan verification.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-7450-647
| | | | - Robert Czarnomysy
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilińskiego 1, 15-089 Bialystok, Poland; (R.C.); (K.B.)
| | - Karol Rogowski
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland;
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilińskiego 1, 15-089 Bialystok, Poland; (R.C.); (K.B.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland;
| |
Collapse
|