1
|
Hunter CD, Cairo CW. Detection Strategies for Sialic Acid and Sialoglycoconjugates. Chembiochem 2024; 25:e202400402. [PMID: 39444251 DOI: 10.1002/cbic.202400402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Indexed: 10/25/2024]
Abstract
Glycoconjugates are a vast class of biomolecules implicated in biological processes important for human health and disease. The structural complexity of glycoconjugates remains a challenge to deciphering their precise biological roles and for their development as biomarkers and therapeutics. Human glycoconjugates on the outside of the cell are modified with sialic (neuraminic) acid residues at their termini. The enzymes that install sialic acids are sialyltransferases (SiaTs), a family of 20 different isoenzymes. The removal and degradation of sialic acids is mediated by neuraminidase (NEU; sialidase) enzymes, of which there are four isoenzymes. In this review, we discuss chemical and biochemical approaches for the detection and analysis of sialoglycoconjugate (SGC) structures and their enzymatic products. The most common methods include affinity probes and synthetic substrates. Fluorogenic and radiolabelled substrates are also important tools for many applications, including screening for enzyme inhibitors. Strategies that give insight into the native substrate-specificity of enzymes that regulate SGCs (SiaT & NEU) are necessary to improve our understanding of the role of sialic acid metabolism in health and disease.
Collapse
Affiliation(s)
- Carmanah D Hunter
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Christopher W Cairo
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| |
Collapse
|
2
|
Zhang X, Hao P, Mo J, Wang PY, Wang G, Li L, Zheng XJ, Yuan X, Yao W, Jin N, Li C, Ye XS. Local and Noninvasive Glyco-Virus Checkpoint Nanoblockades Restrict Sialylation for Prolonged Broad-Spectrum Epidemic Virus Therapy. ACS NANO 2024; 18:32910-32923. [PMID: 39536146 DOI: 10.1021/acsnano.4c12434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has driven major advances in virus research. The role of glycans in viral infection has been revealed, with research demonstrating that terminal sialic acids are key receptors during viral attachment and infection into host cells. However, there is an urgent demand for universal tools to study the mechanism of sialic acids in viral infections, as well as to develop therapeutic agents against epidemic viruses through the downregulation of terminal sialic acid residues on glycans acting as a glyco-virus checkpoint to accelerate virus clearance. In this study, we developed a robust sialic acids blockade tool termed local and noninvasive glyco-virus checkpoint nanoblockades (LONG NBs), which blocked cell surface sialic acids by endogenously and continuously inhibiting the de novo sialic acids biosynthesis pathway. Furthermore, LONG NBs could accurately characterize the sialic acid-dependent profiles of multiple virus variants and protected the host against partial SARS-CoV-2, rotavirus, and influenza A virus infections after local and noninvasive administration. Our results suggest that LONG NBs represent a promising tool to facilitate in-depth research on the mechanism of viral infection, and serve as a broad-spectrum protectant against existing and emerging viral variants via glyco-virus checkpoint blockade.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Pengfei Hao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun 130000, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Juan Mo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Peng-Yu Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Guoqing Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun 130000, China
| | - Letian Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Wenlong Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Chang Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| |
Collapse
|
3
|
Zhang X, Li ZY, Xiao JH, Hao PF, Mo J, Zheng XJ, Geng YQ, Ye XS. Sialic Acids Blockade-Based Chemo-Immunotherapy Featuring Cancer Cell Chemosensitivity and Antitumor Immune Response Synergies. Adv Healthc Mater 2024; 13:e2401649. [PMID: 38938121 DOI: 10.1002/adhm.202401649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Immune checkpoint blockade (ICB) has significantly improved the prognosis of patients with cancer, although the majority of such patients achieve low response rates; consequently, new therapeutic approaches are urgently needed. The upregulation of sialic acid-containing glycans is a common characteristic of cancer-related glycosylation, which drives disease progression and immune escape via numerous pathways. Herein, the development of self-assembled core-shell nanoscale coordination polymer nanoparticles loaded with a sialyltransferase inhibitor, referred to as NCP-STI which effectively stripped diverse sialoglycans from cancer cells, providing an antibody-independent pattern to disrupt the emerging Siglec-sialic acid glyco-immune checkpoint is reported. Furthermore, NCP-STI inhibits sialylation of the concentrated nucleoside transporter 1 (CNT1), promotes the intracellular accumulation of anticancer agent gemcitabine (Gem), and enhances Gem-induced immunogenic cell death (ICD). As a result, the combination of NCP-STI and Gem (NCP-STI/Gem) evokes a robust antitumor immune response and exhibits superior efficacy in restraining the growth of multiple murine tumors and pulmonary metastasis. Collectively, the findings demonstrate a novel form of small molecule-based chemo-immunotherapy approach which features sialic acids blockade that enables cooperative effects of cancer cell chemosensitivity and antitumor immune responses for cancer treatment.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Zi-Yi Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Jia-Heng Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Peng-Fei Hao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Juan Mo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Yi-Qun Geng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing, 100191, China
| |
Collapse
|
4
|
Mukherjee MM, Biesbrock D, Abramowitz LK, Pavan M, Kumar B, Walter PJ, Azadi P, Jacobson KA, Hanover JA. Selective bioorthogonal probe for N-glycan hybrid structures. Nat Chem Biol 2024:10.1038/s41589-024-01756-5. [PMID: 39468349 DOI: 10.1038/s41589-024-01756-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/17/2024] [Indexed: 10/30/2024]
Abstract
Metabolic incorporation of chemically tagged monosaccharides is a facile means of tagging cellular glycoproteins and glycolipids. However, since the monosaccharide precursors are often shared by several pathways, selectivity has been difficult to attain. For example, N-linked glycosylation is a chemically complex and ubiquitous posttranslational modification, with three distinct classes of GlcNAc-containing N-glycan structures: oligomannose, hybrid and complex. Here we describe the synthesis of 1,3-Pr2-6-OTs GlcNAlk (MM-JH-1) as a next-generation metabolic chemical reporter for the selective labeling of hybrid N-glycan structures. We first developed a general strategy for defining the selectivity of labeling with chemically tagged monosaccharides. We then applied this approach to establish that MM-JH-1 is selectively incorporated into hybrid N-glycans. Using this metabolic chemical reporter as a detection tool, we performed imaging and fractionation to define features of the intracellular localization and trafficking of target proteins bearing hybrid N-glycan structures.
Collapse
Affiliation(s)
- Mana Mohan Mukherjee
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Devin Biesbrock
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Lara K Abramowitz
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Matteo Pavan
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Peter J Walter
- Clinical Mass Spectrometry Core, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, Athens, GA, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - John A Hanover
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Idiago-López J, Ferreira D, Asín L, Moros M, Armenia I, Grazú V, Fernandes AR, de la Fuente JM, Baptista PV, Fratila RM. Membrane-localized magnetic hyperthermia promotes intracellular delivery of cell-impermeant probes. NANOSCALE 2024; 16:15176-15195. [PMID: 39052238 DOI: 10.1039/d4nr01955e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
In this work, we report the disruptive use of membrane-localized magnetic hyperthermia to promote the internalization of cell-impermeant probes. Under an alternating magnetic field, magnetic nanoparticles (MNPs) immobilized on the cell membrane via bioorthogonal click chemistry act as nanoheaters and lead to the thermal disruption of the plasma membrane, which can be used for internalization of different types of molecules, such as small fluorescent probes and nucleic acids. Noteworthily, no cell death, oxidative stress and alterations of the cell cycle are detected after the thermal stimulus, although cells are able to sense and respond to the thermal stimulus through the expression of different types of heat shock proteins (HSPs). Finally, we demonstrate the utility of this approach for the transfection of cells with a small interference RNA (siRNA), revealing a similar efficacy to a standard transfection method based on the use of cationic lipid-based reagents (such as Lipofectamine), but with lower cell toxicity. These results open the possibility of developing new procedures for "opening and closing" cellular membranes with minimal disturbance of cellular integrity. This on-demand modification of cell membrane permeability could allow the direct intracellular delivery of biologically relevant (bio)molecules, drugs and nanomaterials, thus overcoming traditional endocytosis pathways and avoiding endosomal entrapment.
Collapse
Affiliation(s)
- Javier Idiago-López
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Daniela Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Laura Asín
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Moros
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Jesús M de la Fuente
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, 50009, Zaragoza, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departamento de Química Orgánica, Facultad de Ciencias, C/Pedro Cerbuna 12, 50009, Zaragoza, Spain
| |
Collapse
|
6
|
Zhang J, Terreni M, Liu F, Sollogoub M, Zhang Y. Ganglioside GM3-based anticancer vaccines: Reviewing the mechanism and current strategies. Biomed Pharmacother 2024; 176:116824. [PMID: 38820973 DOI: 10.1016/j.biopha.2024.116824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Ganglioside GM3 is one of the most common membrane-bound glycosphingolipids. The over-expression of GM3 on tumor cells makes it defined as a tumor-associated carbohydrate antigen (TACA). The specific expression property in cancers, especially in melanoma, make it become an important target to develop anticancer vaccines or immunotherapies. However, in the manner akin to most TACAs, GM3 is an autoantigen facing with problems of low immunogenicity and easily inducing immunotolerance, which means itself only cannot elicit a powerful enough immune response to prevent or treat cancer. With a comparative understanding of the mechanisms that how immune system responses to the carbohydrate vaccines, this review summarizes the studies on the recent efforts to development GM3-based anticancer vaccines.
Collapse
Affiliation(s)
- Jiaxu Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Fang Liu
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Matthieu Sollogoub
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Yongmin Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
7
|
Li W, Guo J, Hobson EC, Xue X, Li Q, Fu J, Deng CX, Guo Z. Metabolic-Glycoengineering-Enabled Molecularly Specific Acoustic Tweezing Cytometry for Targeted Mechanical Stimulation of Cell Surface Sialoglycans. Angew Chem Int Ed Engl 2024; 63:e202401921. [PMID: 38498603 PMCID: PMC11073901 DOI: 10.1002/anie.202401921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/20/2024]
Abstract
In this study, we developed a novel type of dibenzocyclooctyne (DBCO)-functionalized microbubbles (MBs) and validated their attachment to azide-labelled sialoglycans on human pluripotent stem cells (hPSCs) generated by metabolic glycoengineering (MGE). This enabled the application of mechanical forces to sialoglycans on hPSCs through molecularly specific acoustic tweezing cytometry (mATC), that is, displacing sialoglycan-anchored MBs using ultrasound (US). It was shown that subjected to the acoustic radiation forces of US pulses, sialoglycan-anchored MBs exhibited significantly larger displacements and faster, more complete recovery after each pulse than integrin-anchored MBs, indicating that sialoglycans are more stretchable and elastic than integrins on hPSCs in response to mechanical force. Furthermore, stimulating sialoglycans on hPSCs using mATC reduced stage-specific embryonic antigen-3 (SSEA-3) and GD3 expression but not OCT4 and SOX2 nuclear localization. Conversely, stimulating integrins decreased OCT4 nuclear localization but not SSEA-3 and GD3 expression, suggesting that mechanically stimulating sialoglycans and integrins initiated distinctive mechanoresponses during the early stages of hPSC differentiation. Taken together, these results demonstrated that MGE-enabled mATC uncovered not only different mechanical properties of sialoglycans on hPSCs and integrins but also their different mechanoregulatory impacts on hPSC differentiation, validating MGE-based mATC as a new, powerful tool for investigating the roles of glycans and other cell surface biomolecules in mechanotransduction.
Collapse
Affiliation(s)
- Weiping Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiatong Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Eric C. Hobson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qingjiang Li
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- Department of Chemistry, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Jianping Fu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cheri X. Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
8
|
Xu J, Sun Y, You Y, Zhang Y, Huang D, Zhou S, Liu Y, Tong S, Ma F, Song Q, Dai C, Li S, Lei J, Wang Z, Gao X, Chen J. Bioorthogonal microglia-inspired mesenchymal stem cell bioengineering system creates livable niches for enhancing ischemic stroke recovery via the hormesis. Acta Pharm Sin B 2024; 14:1412-1427. [PMID: 38486994 PMCID: PMC10935060 DOI: 10.1016/j.apsb.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/11/2023] [Accepted: 09/28/2023] [Indexed: 03/17/2024] Open
Abstract
Mesenchymal stem cells (MSCs) experience substantial viability issues in the stroke infarct region, limiting their therapeutic efficacy and clinical translation. High levels of deadly reactive oxygen radicals (ROS) and proinflammatory cytokines (PC) in the infarct milieu kill transplanted MSCs, whereas low levels of beneficial ROS and PC stimulate and improve engrafted MSCs' viability. Based on the intrinsic hormesis effects in cellular biology, we built a microglia-inspired MSC bioengineering system to transform detrimental high-level ROS and PC into vitality enhancers for strengthening MSC therapy. This system is achieved by bioorthogonally arming metabolic glycoengineered MSCs with microglial membrane-coated nanoparticles and an antioxidative extracellular protective layer. In this system, extracellular ROS-scavenging and PC-absorbing layers effectively buffer the deleterious effects and establish a micro-livable niche at the level of a single MSC for transplantation. Meanwhile, the infarct's inanimate milieu is transformed at the tissue level into a new living niche to facilitate healing. The engineered MSCs achieved viability five times higher than natural MSCs at seven days after transplantation and exhibited a superior therapeutic effect for stroke recovery up to 28 days. This vitality-augmented system demonstrates the potential to accelerate the clinical translation of MSC treatment and boost stroke recovery.
Collapse
Affiliation(s)
- Jianpei Xu
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yinzhe Sun
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yang You
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence & Department of Neurology, Fudan University, Shanghai 201203, China
| | - Dan Huang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yipu Liu
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shiqiang Tong
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Fenfen Ma
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chengxiang Dai
- Daxing Research Institute, University of Science and Technology Beijing, Biomedical Industry Base, Zhongguancun Science and Technology Park, Beijing 102600, China
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Suke Li
- Daxing Research Institute, University of Science and Technology Beijing, Biomedical Industry Base, Zhongguancun Science and Technology Park, Beijing 102600, China
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Jigang Lei
- Daxing Research Institute, University of Science and Technology Beijing, Biomedical Industry Base, Zhongguancun Science and Technology Park, Beijing 102600, China
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Zhihua Wang
- Department of Emergency, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
9
|
Li Y, Wang H, Chen Y, Ding L, Ju H. In Situ Glycan Analysis and Editing in Living Systems. JACS AU 2024; 4:384-401. [PMID: 38425935 PMCID: PMC10900212 DOI: 10.1021/jacsau.3c00717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 03/02/2024]
Abstract
Besides proteins and nucleic acids, carbohydrates are also ubiquitous building blocks of living systems. Approximately 70% of mammalian proteins are glycosylated. Glycans not only provide structural support for living systems but also act as crucial regulators of cellular functions. As a result, they are considered essential pieces of the life science puzzle. However, research on glycans has lagged far behind that on proteins and nucleic acids. The main reason is that glycans are not direct products of gene coding, and their synthesis is nontemplated. In addition, the diversity of monosaccharide species and their linkage patterns contribute to the complexity of the glycan structures, which is the molecular basis for their diverse functions. Research in glycobiology is extremely challenging, especially for the in situ elucidation of glycan structures and functions. There is an urgent need to develop highly specific glycan labeling tools and imaging methods and devise glycan editing strategies. This Perspective focuses on the challenges of in situ analysis of glycans in living systems at three spatial levels (i.e., cell, tissue, and in vivo) and highlights recent advances and directions in glycan labeling, imaging, and editing tools. We believe that examining the current development landscape and the existing bottlenecks can drive the evolution of in situ glycan analysis and intervention strategies and provide glycan-based insights for clinical diagnosis and therapeutics.
Collapse
Affiliation(s)
- Yiran Li
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Haiqi Wang
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Yunlong Chen
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| | - Lin Ding
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
- Chemistry
and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing
University, Nanjing 210023, China
| |
Collapse
|
10
|
Alghazali R, Nugud A, El-Serafi A. Glycan Modifications as Regulators of Stem Cell Fate. BIOLOGY 2024; 13:76. [PMID: 38392295 PMCID: PMC10886185 DOI: 10.3390/biology13020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
Glycosylation is a process where proteins or lipids are modified with glycans. The presence of glycans determines the structure, stability, and localization of glycoproteins, thereby impacting various biological processes, including embryogenesis, intercellular communication, and disease progression. Glycans can influence stem cell behavior by modulating signaling molecules that govern the critical aspects of self-renewal and differentiation. Furthermore, being located at the cell surface, glycans are utilized as markers for stem cell pluripotency and differentiation state determination. This review aims to provide a comprehensive overview of the current literature, focusing on the effect of glycans on stem cells with a reflection on the application of synthetic glycans in directing stem cell differentiation. Additionally, this review will serve as a primer for researchers seeking a deeper understanding of how synthetic glycans can be used to control stem cell differentiation, which may help establish new approaches to guide stem cell differentiation into specific lineages. Ultimately, this knowledge can facilitate the identification of efficient strategies for advancing stem cell-based therapeutic interventions.
Collapse
Affiliation(s)
- Raghad Alghazali
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
| | - Ahmed Nugud
- Clinical Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Gastroenterology, Hepatology & Nutrition, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
11
|
Liu Y, Wang H. Biomarkers and targeted therapy for cancer stem cells. Trends Pharmacol Sci 2024; 45:56-66. [PMID: 38071088 PMCID: PMC10842814 DOI: 10.1016/j.tips.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 01/07/2024]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cancer cells with capabilities of self-renewal, differentiation, and tumorigenicity, and play a critical role in driving tumor heterogeneity that evolves insensitivity to therapeutics. For these reasons, extensive efforts have been made to identify and target CSCs to potentially improve the antitumor efficacy of therapeutics. While progress has been made to uncover certain CSC-associated biomarkers, the identification of CSC-specific markers, especially the targetable ones, remains a significant challenge. Here we provide an overview of the unique signaling and metabolic pathways of CSCs, summarize existing CSC biomarkers and CSC-targeted therapies, and discuss strategies to further differentiate CSCs from non-stem cancer cells and healthy cells for the development of enhanced CSC-targeted therapies.
Collapse
Affiliation(s)
- Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Cancer Center at Illinois (CCIL), Urbana, IL 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
12
|
Min Q, Ji X. Bioorthogonal Bond Cleavage Chemistry for On-demand Prodrug Activation: Opportunities and Challenges. J Med Chem 2023; 66:16546-16567. [PMID: 38085596 DOI: 10.1021/acs.jmedchem.3c01459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Time- and space-resolved drug delivery is highly demanded for cancer treatment, which, however, can barely be achieved with a traditional prodrug strategy. In recent years, the prodrug strategy based on a bioorthogonal bond cleavage chemistry has emerged with the advantages of high temporospatial resolution over drug activation and homogeneous activation irrespective of individual heterogeneity. In the past five years, tremendous progress has been witnessed in this field with one such bioorthogonal prodrug entering Phase II clinical trials. This Perspective aims to highlight these new advances (2019-2023) and critically discuss their pros and cons. In addition, the remaining challenges and potential strategic directions for future progress will also be included.
Collapse
Affiliation(s)
- Qingqiang Min
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Xingyue Ji
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
13
|
Mukherjee MM, Bond MR, Abramowitz LK, Biesbrock D, Woodroofe CC, Kim EJ, Swenson RE, Hanover JA. Tools and tactics to define specificity of metabolic chemical reporters. Front Mol Biosci 2023; 10:1286690. [PMID: 38143802 PMCID: PMC10740162 DOI: 10.3389/fmolb.2023.1286690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
Metabolic chemical reporters (MCRs) provide easily accessible means to study glycans in their native environments. However, because monosaccharide precursors are shared by many glycosylation pathways, selective incorporation has been difficult to attain. Here, a strategy for defining the selectivity and enzymatic incorporation of an MCR is presented. Performing β-elimination to interrogate O-linked sugars and using commercially available glycosidases and glycosyltransferase inhibitors, we probed the specificity of widely used azide (Ac4GalNAz) and alkyne (Ac4GalNAlk and Ac4GlcNAlk) sugar derivatives. Following the outlined strategy, we provide a semiquantitative assessment of the specific and non-specific incorporation of this bioorthogonal sugar (Ac4GalNAz) into numerous N- and O-linked glycosylation pathways. This approach should be generally applicable to other MCRs to define the extent of incorporation into the various glycan species.
Collapse
Affiliation(s)
- Mana Mohan Mukherjee
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michelle R. Bond
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lara K. Abramowitz
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Devin Biesbrock
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Carolyn C. Woodroofe
- Frederick National Laboratory for Cancer Research, National Cancer Institute, Fredrick, MD, United States
| | - Eun Ju Kim
- Department of Chemistry Education, Daegu University, Gyeongsan-si, South Korea
| | - Rolf E. Swenson
- Department of Chemistry Education, Daegu University, Gyeongsan-si, South Korea
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
14
|
Mirkale K, Jain SK, Oviya TS, Mahalingam S. Optomicrofluidic detection of cancer cells in peripheral blood via metabolic glycoengineering. LAB ON A CHIP 2023; 23:5151-5164. [PMID: 37955355 DOI: 10.1039/d3lc00678f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The currently existing label-based techniques for the detection of circulating tumor cells (CTCs) target natural surface proteins of cells and are therefore applicable to only limited cancer cell types. We report optomicrofluidic detection of cancer cells in the pool of peripheral blood mononuclear cells (PBMCs) by exploiting the difference in their cell metabolism. We employ metabolic glycoengineering as a click chemistry tool for tagging cells that yields several fold-higher fluorescence signals from cancer cells compared to that from PBMCs. The effects of concentrations of the tagging compounds and cell incubation time on the fluorescence signal intensity are studied. The tagged cells were encapsulated in droplets ensuring that cells enter the detection region two-dimensionally focused in single-file and optically detected with a high detection efficiency and low coefficient of variation of the signals. The metabolic tagging approach showed a significantly higher tagging efficiency and average fluorescence signal compared to the well-established and widely adopted anti-EpCAM-FITC-based tagging. We demonstrated the detection of three different cancer cell lines - EpCAM-negative cervical cancer cell, HeLa, weakly EpCAM positive, and triple-negative breast cancer cell, MDA-MB-231, and strongly EpCAM positive breast cancer cell, MCF7, highlighting that the proposed technique is independent of naturally occurring cell surface proteins and widely applicable. The metabolically tagged and optically detected cells were successfully recultured, proving the compatibility of the proposed technique with downstream assays. The proposed technique is then utilised for the detection of CTCs in metastatic cancer patients' blood. The current work provides a new strategy for detecting cancer cells in the blood that can find potential applications in both fundamental research and clinical studies involving CTCs as well as in single-cell sequencing.
Collapse
Affiliation(s)
- K Mirkale
- Micro Nano Bio Fluidics Unit, Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai-600036, Tamilnadu, India.
| | - S K Jain
- Micro Nano Bio Fluidics Unit, Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai-600036, Tamilnadu, India.
| | - T S Oviya
- Micro Nano Bio Fluidics Unit, Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai-600036, Tamilnadu, India.
| | - S Mahalingam
- Laboratory of Molecular Cell Biology, National Cancer Tissue Biobank, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai-600036, India
| |
Collapse
|
15
|
Almeida‐Pinto J, Lagarto MR, Lavrador P, Mano JF, Gaspar VM. Cell Surface Engineering Tools for Programming Living Assemblies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304040. [PMID: 37823678 PMCID: PMC10700290 DOI: 10.1002/advs.202304040] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/10/2023] [Indexed: 10/13/2023]
Abstract
Breakthroughs in precision cell surface engineering tools are supporting the rapid development of programmable living assemblies with valuable features for tackling complex biological problems. Herein, the authors overview the most recent technological advances in chemically- and biologically-driven toolboxes for engineering mammalian cell surfaces and triggering their assembly into living architectures. A particular focus is given to surface engineering technologies for enabling biomimetic cell-cell social interactions and multicellular cell-sorting events. Further advancements in cell surface modification technologies may expand the currently available bioengineering toolset and unlock a new generation of personalized cell therapeutics with clinically relevant biofunctionalities. The combination of state-of-the-art cell surface modifications with advanced biofabrication technologies is envisioned to contribute toward generating living materials with increasing tissue/organ-mimetic bioactivities and therapeutic potential.
Collapse
Affiliation(s)
- José Almeida‐Pinto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Matilde R. Lagarto
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Pedro Lavrador
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - João F. Mano
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| | - Vítor M. Gaspar
- Department of ChemistryCICECO‐Aveiro Institute of Materials University of Aveiro Campus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
16
|
Le B, Zhu K, Brown C, Reid B, Cressman A, Zhao M, Fierro FA. Reducing Sialylation Enhances Electrotaxis of Corneal Epithelial Cells. Int J Mol Sci 2023; 24:14327. [PMID: 37762630 PMCID: PMC10531958 DOI: 10.3390/ijms241814327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Corneal wound healing is a complex biological process that integrates a host of different signals to coordinate cell behavior. Upon wounding, there is the generation of an endogenous wound electric field that serves as a powerful cue to guide cell migration. Concurrently, the corneal epithelium reduces sialylated glycoforms, suggesting that sialylation plays an important role during electrotaxis. Here, we show that pretreating human telomerase-immortalized corneal epithelial (hTCEpi) cells with a sialyltransferase inhibitor, P-3FAX-Neu5Ac (3F-Neu5Ac), improves electrotaxis by enhancing directionality, but not speed. This was recapitulated using Kifunensine, which inhibits cleavage of mannoses and therefore precludes sialylation on N-glycans. We also identified that 3F-Neu5Ac enhanced the responsiveness of the hTCEpi cell population to the electric field and that pretreated hTCEpi cells showed increased directionality even at low voltages. Furthermore, when we increased sialylation using N-azidoacetylmannosamine-tetraacylated (Ac4ManNAz), hTCEpi cells showed a decrease in both speed and directionality. Importantly, pretreating enucleated eyes with 3F-Neu5Ac significantly improved re-epithelialization in an ex vivo model of a corneal injury. Finally, we show that in hTCEpi cells, sialylation is increased by growth factor deprivation and reduced by PDGF-BB. Taken together, our results suggest that during corneal wound healing, reduced sialylated glycoforms enhance electrotaxis and re-epithelialization, potentially opening new avenues to promote corneal wound healing.
Collapse
Affiliation(s)
- Bryan Le
- Department of Ophthalmology, University of California, Davis, CA 95616, USA; (B.L.); (M.Z.)
| | - Kan Zhu
- Department of Ophthalmology, University of California, Davis, CA 95616, USA; (B.L.); (M.Z.)
| | - Chelsea Brown
- Department of Ophthalmology, University of California, Davis, CA 95616, USA; (B.L.); (M.Z.)
| | - Brian Reid
- Department of Ophthalmology, University of California, Davis, CA 95616, USA; (B.L.); (M.Z.)
| | - Amin Cressman
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95817, USA
| | - Min Zhao
- Department of Ophthalmology, University of California, Davis, CA 95616, USA; (B.L.); (M.Z.)
| | - Fernando A. Fierro
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95817, USA
| |
Collapse
|
17
|
Weiser-Fuchs MT, Maggauer E, van Poppel MNM, Csapo B, Desoye G, Köfeler HC, Groselj-Strele A, Trajanoski S, Fluhr H, Obermayer-Pietsch B, Jantscher-Krenn E. Human Milk Oligosaccharides in Maternal Serum Respond to Oral Glucose Load and Are Associated with Insulin Sensitivity. Nutrients 2023; 15:4042. [PMID: 37764825 PMCID: PMC10534497 DOI: 10.3390/nu15184042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: Pregnancy presents a challenge to maternal glucose homeostasis; suboptimal adaptations can lead to gestational diabetes mellitus (GDM). Human milk oligosaccharides (HMOs) circulate in maternal blood in pregnancy and are altered with GDM, suggesting influence of glucose homeostasis on HMOs. We thus assessed the HMO response to glucose load during an oral glucose tolerance test (OGTT) and investigated HMO associations with glucose tolerance/insulin sensitivity in healthy pregnant women. (2) Methods: Serum of 99 women, collected at 0 h, 1 h and 2 h during a 75 g OGTT at 24-28 gestational weeks was analyzed for HMOs (2'FL, 3'SLN, LDFT, 3'SL) by HPLC; plasma glucose, insulin and C-peptide were analyzed by standard biochemistry methods. (3) Results: Serum 3'SL concentrations significantly increased from fasting to 1 h after glucose load, while concentrations of the other HMOs were unaltered. Higher 3'SL at all OGTT time points was associated with a generally more diabetogenic profile, with higher hepatic insulin resistance (HOMA-IR), lower insulin sensitivity (Matsuda index) and higher insulin secretion (C-peptide index 1). (4) Conclusions: Rapid increase in serum 3'SL post-oral glucose load (fasted-fed transition) indicates utilization of plasma glucose, potentially for sialylation of lactose. Associations of sialylated HMOs with a more diabetogenic profile suggest sustained adaptations to impaired glucose homeostasis in pregnancy. Underlying mechanisms or potential consequences of observed HMO changes remain to be elucidated.
Collapse
Affiliation(s)
- Marie-Therese Weiser-Fuchs
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, 8036 Graz, Austria
| | - Elena Maggauer
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
| | - Mireille N. M. van Poppel
- Institute of Human Movement Science, Sport and Health, University of Graz, 8010 Graz, Austria;
- BioTechMed, 8010 Graz, Austria;
| | - Bence Csapo
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, 8036 Graz, Austria
| | - Harald C. Köfeler
- BioTechMed, 8010 Graz, Austria;
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Groselj-Strele
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, 8036 Graz, Austria; (A.G.-S.); (S.T.)
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Center for Medical Research, Medical University of Graz, 8036 Graz, Austria; (A.G.-S.); (S.T.)
| | - Herbert Fluhr
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, 8036 Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria;
- Department of Obstetrics and Gynecology, Endocrinology Lab Platform, 8036 Graz, Austria
| | - Evelyn Jantscher-Krenn
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (M.-T.W.-F.); (G.D.); (E.J.-K.)
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, 8036 Graz, Austria
- BioTechMed, 8010 Graz, Austria;
| |
Collapse
|
18
|
Zhou H, Zhang Y, Pei P, Shen W, Yi X, Yang K. Liposome-anchored mesenchymal stem cells for radiation pneumonia/fibrosis treatment. Biomaterials 2023; 300:122202. [PMID: 37336116 DOI: 10.1016/j.biomaterials.2023.122202] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
The effectiveness of mesenchymal stem cells (MSCs) on inflammation-related disease is limited and the pharmaceutical preparation that was used to enhance the efficacy of MSCs cannot reach the diseased tissue in large quantities. Herein, antioxidant liposome (Lipo-OPC) is designed to anchor onto the surface of MSCs membrane via click chemical reaction (MSC-Lipo-OPC) without affecting the viability and physiological characteristics of MSCs, thus allowing efficient accumulation of MSC-Lipo-OPC in X-ray irradiated lung sites. More importantly, MSC-Lipo-OPC promotes the change of the quantity and polarity of innate immunocytes, mainly including neutrophils, macrophages and Tregs, in favor of anti-inflammatory, finally preventing the formation of radioactive pulmonary fibrosis. Therefore, it could enhance the treatment outcome of both of MSCs and drugs to radiation-induced lung injury via modifying the drug-loaded nanoparticle on the surface of MSCs membrane, further promoting the application of MSCs in radiation damage and protection.
Collapse
Affiliation(s)
- Hailin Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Wenhao Shen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuan Yi
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, Jiangsu, 226001, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
19
|
Mitry MMA, Boateng SY, Greco F, Osborn HMI. Bioorthogonal activation of prodrugs, for the potential treatment of breast cancer, using the Staudinger reaction. RSC Med Chem 2023; 14:1537-1548. [PMID: 37593579 PMCID: PMC10429771 DOI: 10.1039/d3md00137g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/03/2023] [Indexed: 08/19/2023] Open
Abstract
Selective prodrug activation at a tumor site is crucial to maximise the efficiency of chemotherapy approaches and minimise side effects due to off-site activation. In this paper, a new prodrug activation strategy is reported based on the bioorthogonal Staudinger reaction. The feasibility of this prodrug activation strategy was initially demonstrated using 9-azido sialic acid 4 as a trigger and two novel triphenylphosphine-modified N-mustard-PRO 10 and doxorubicin-PRO 12 prodrugs in an HPLC-monitored release study. Then, the azide reporter group was introduced on cancer cells' surfaces through metabolic glycoengineering of sialic acid-rich surface glycans using azide-modified monosaccharides (9-azido sialic acid 4, tetra-O-acetylated-9-azido sialic acid 5 and tetra-O-acetyl azidomannosamine). Next, the N-mustard-PRO 10 and doxorubicin-PRO 12 prodrugs were employed in vitro with the bioengineered cells, and activation of the prodrugs, which allowed selective release of the cytotoxic moiety at the tumour cell, was assessed. Release of the parent drugs from the prodrugs was shown to be dependent on the level of metabolic labelling, where tetra-O-acetyl azidomannosamine allowed the highest level of azide reporter generation in tumor cells and led to full recovery of the parent cytotoxic drug's potency. The selectivity of azide expression on breast cancer MCF-7 cells versus normal fibroblast L929 cells was also probed, with the 9-azido sialic acid and tetra-O-acetylated-9-azido sialic acid showing ∼17-fold higher azide expression on the former. Taken together, these data demonstrate the feasibility of the Staudinger reaction for selective activation of prodrugs targeted to the MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Madonna M A Mitry
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
- Dept. of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University Cairo 11566 Egypt
| | - Samuel Y Boateng
- School of Biological Sciences, University of Reading Whiteknights Reading RG6 6ES UK
| | - Francesca Greco
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
| | - Helen M I Osborn
- Reading School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
| |
Collapse
|
20
|
Mukherjee MM, Abramowitz LK, Kumar B, Azadi P, Hanover JA. Selective bioorthogonal probe for N-glycan hybrid structures. RESEARCH SQUARE 2023:rs.3.rs-3093724. [PMID: 37577573 PMCID: PMC10418551 DOI: 10.21203/rs.3.rs-3093724/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Metabolic incorporation of chemically tagged monosaccharides is a facile means of labelling cellular glycoprotein and glycolipids. Yet, since the monosaccharide precursors are often shared by several pathways, selectivity has been difficult to attain. For example, N-linked glycosylation is a chemically complex, and ubiquitous post translational modification with three distinct classes of GlcNAc-containing N-glycan structures: oligomannose, hybrid, and complex. Here we describe synthesis of 1,3-Pr2-6-OTs GlcNAlk as a next generation metabolic chemical reporter (MCR) for the specific labeling of hybrid N-glycan structures. We first developed a general strategy for defining the selectivity of labelling with chemically tagged monosaccharides. We then applied this approach to establish that 1,3-Pr2-6-OTs GlcNAlk is specifically incorporated into hybrid N-glycans. Using this MCR as a detection tool, we carried out imaging experiments to define the intracellular localization and trafficking of target proteins bearing hybrid N-glycan structures.
Collapse
Affiliation(s)
- Mana Mohan Mukherjee
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD
| | - Lara K Abramowitz
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - John A Hanover
- Laboratory of Cell and Molecular Biology, NIDDK, National Institutes of Health, Bethesda, MD
| |
Collapse
|
21
|
Du J, Wang Z, Liu X, Hu C, Yarema KJ, Jia X. Improving Schwann Cell Differentiation from Human Adipose Stem Cells with Metabolic Glycoengineering. Cells 2023; 12:1190. [PMID: 37190099 PMCID: PMC10136940 DOI: 10.3390/cells12081190] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Schwann cells (SCs) are myelinating cells that promote peripheral nerve regeneration. When nerve lesions form, SCs are destroyed, ultimately hindering nerve repair. The difficulty in treating nerve repair is exacerbated due to SC's limited and slow expansion capacity. Therapeutic use of adipose-derived stem cells (ASCs) is emerging in combating peripheral nerve injury due to these cells' SC differentiation capability and can be harvested easily in large numbers. Despite ASC's therapeutic potential, their transdifferentiation period typically takes more than two weeks. In this study, we demonstrate that metabolic glycoengineering (MGE) technology enhances ASC differentiation into SCs. Specifically, the sugar analog Ac5ManNTProp (TProp), which modulates cell surface sialylation, significantly improved ASC differentiation with upregulated SC protein S100β and p75NGFR expression and elevated the neurotrophic factors nerve growth factor beta (NGFβ) and glial cell-line-derived neurotrophic factor (GDNF). TProp treatment remarkably reduced the SC transdifferentiation period from about two weeks to two days in vitro, which has the potential to improve neuronal regeneration and facilitate future use of ASCs in regenerative medicine.
Collapse
Affiliation(s)
- Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MST 823, Baltimore, MD 21201, USA
| | - Zihui Wang
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MST 823, Baltimore, MD 21201, USA
| | - Xiao Liu
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MST 823, Baltimore, MD 21201, USA
| | - Cecilia Hu
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MST 823, Baltimore, MD 21201, USA
| | - Kevin J. Yarema
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MST 823, Baltimore, MD 21201, USA
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
22
|
Metabolic Glycoengineering: A Promising Strategy to Remodel Microenvironments for Regenerative Therapy. Stem Cells Int 2023; 2023:1655750. [PMID: 36814525 PMCID: PMC9940976 DOI: 10.1155/2023/1655750] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 09/27/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023] Open
Abstract
Cell-based regenerative therapy utilizes the differentiation potential of stem cells to rejuvenate tissues. But the dynamic fate of stem cells is calling for precise control to optimize their therapeutic efficiency. Stem cell fate is regulated by specific conditions called "microenvironments." Among the various factors in the microenvironment, the cell-surface glycan acts as a mediator of cell-matrix and cell-cell interactions and manipulates the behavior of cells. Herein, metabolic glycoengineering (MGE) is an easy but powerful technology for remodeling the structure of glycan. By presenting unnatural glycans on the surface, MGE provides us an opportunity to reshape the microenvironment and evoke desired cellular responses. In this review, we firstly focused on the determining role of glycans on cellular activity; then, we introduced how MGE influences glycosylation and subsequently affects cell fate; at last, we outlined the application of MGE in regenerative therapy, especially in the musculoskeletal system, and the future direction of MGE is discussed.
Collapse
|
23
|
Saeui CT, Shah SR, Fernandez-Gil BI, Zhang C, Agatemor C, Dammen-Brower K, Mathew MP, Buettner M, Gowda P, Khare P, Otamendi-Lopez A, Yang S, Zhang H, Le A, Quinoñes-Hinojosa A, Yarema KJ. Anticancer Properties of Hexosamine Analogs Designed to Attenuate Metabolic Flux through the Hexosamine Biosynthetic Pathway. ACS Chem Biol 2023; 18:151-165. [PMID: 36626752 DOI: 10.1021/acschembio.2c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Altered cellular metabolism is a hallmark of cancer pathogenesis and progression; for example, a near-universal feature of cancer is increased metabolic flux through the hexosamine biosynthetic pathway (HBP). This pathway produces uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), a potent oncometabolite that drives multiple facets of cancer progression. In this study, we synthesized and evaluated peracetylated hexosamine analogs designed to reduce flux through the HBP. By screening a panel of analogs in pancreatic cancer and glioblastoma multiform (GBM) cells, we identified Ac4Glc2Bz─a benzyl-modified GlcNAc mimetic─as an antiproliferative cancer drug candidate that down-regulated oncogenic metabolites and reduced GBM cell motility at concentrations non-toxic to non-neoplastic cells. More specifically, the growth inhibitory effects of Ac4Glc2Bz were linked to reduced levels of UDP-GlcNAc and concomitant decreases in protein O-GlcNAc modification in both pancreatic cancer and GBM cells. Targeted metabolomics analysis in GBM cells showed that Ac4Glc2Bz disturbed glucose metabolism, amino acid pools, and nucleotide precursor biosynthesis, consistent with reduced proliferation and other anti-oncogenic properties of this analog. Furthermore, Ac4Glc2Bz reduced the invasion, migration, and stemness of GBM cells. Importantly, normal metabolic functions mediated by UDP-GlcNAc were not disrupted in non-neoplastic cells, including maintenance of endogenous levels of O-GlcNAcylation with no global disruption of N-glycan production. Finally, a pilot in vivo study showed that a potential therapeutic window exists where animals tolerated 5- to 10-fold higher levels of Ac4Glc2Bz than projected for in vivo efficacy. Together, these results establish GlcNAc analogs targeting the HBP through salvage mechanisms as a new therapeutic approach to safely normalize an important facet of aberrant glucose metabolism associated with cancer.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | | | - Cissy Zhang
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Christian Agatemor
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Kris Dammen-Brower
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Mohit P Mathew
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Matthew Buettner
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Prateek Gowda
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Pratik Khare
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Shuang Yang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Hui Zhang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Anne Le
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Kevin J Yarema
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
24
|
Mitry MMA, Greco F, Osborn HMI. In Vivo Applications of Bioorthogonal Reactions: Chemistry and Targeting Mechanisms. Chemistry 2023; 29:e202203942. [PMID: 36656616 DOI: 10.1002/chem.202203942] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Bioorthogonal chemistry involves selective biocompatible reactions between functional groups that are not normally present in biology. It has been used to probe biomolecules in living systems, and has advanced biomedical strategies such as diagnostics and therapeutics. In this review, the challenges and opportunities encountered when translating in vitro bioorthogonal approaches to in vivo settings are presented, with a focus on methods to deliver the bioorthogonal reaction components. These methods include metabolic bioengineering, active targeting, passive targeting, and simultaneously used strategies. The suitability of bioorthogonal ligation reactions and bond cleavage reactions for in vivo applications is critically appraised, and practical considerations such as the optimum scheduling regimen in pretargeting approaches are discussed. Finally, we present our own perspectives for this area and identify what, in our view, are the key challenges that must be overcome to maximise the impact of these approaches.
Collapse
Affiliation(s)
- Madonna M A Mitry
- Reading School of Pharmacy, University of Reading Whiteknights, Reading, RG6 6AD, UK.,Department of Pharmaceutical Chemistry Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Francesca Greco
- Reading School of Pharmacy, University of Reading Whiteknights, Reading, RG6 6AD, UK
| | - Helen M I Osborn
- Reading School of Pharmacy, University of Reading Whiteknights, Reading, RG6 6AD, UK
| |
Collapse
|
25
|
Abstract
Adoptive T cell transfer (ACT) therapies suffer from a number of limitations (e.g., poor control of solid tumors), and while combining ACT with cytokine therapy can enhance effectiveness, this also results in significant side effects. Here, we describe a nanotechnology approach to improve the efficacy of ACT therapies by metabolically labeling T cells with unnatural sugar nanoparticles, allowing direct conjugation of antitumor cytokines onto the T cell surface during the manufacturing process. This allows local, concentrated activity of otherwise toxic cytokines. This approach increases T cell infiltration into solid tumors, activates the host immune system toward a Type 1 response, encourages antigen spreading, and improves control of aggressive solid tumors and achieves complete blood cancer regression with otherwise noncurative doses of CAR-T cells. Overall, this method provides an effective and easily integrated approach to the current ACT manufacturing process to increase efficacy in various settings.
Collapse
|
26
|
Wu Z, Zhang H, Yan J, Wei Y, Su J. Engineered biomembrane-derived nanoparticles for nanoscale theranostics. Theranostics 2023; 13:20-39. [PMID: 36593970 PMCID: PMC9800735 DOI: 10.7150/thno.76894] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022] Open
Abstract
Currently, biological membrane-derived nanoparticles (NPs) have shown enormous potential as drug delivery vehicles due to their outstanding biomimetic properties. To make these NPs more adaptive to complex biological systems, some methods have been developed to modify biomembranes and endow them with more functions while preserving their inherent natures. In this review, we introduce five common approaches used for biomembrane decoration: membrane hybridization, the postinsertion method, chemical methods, metabolism engineering and gene engineering. These methods can functionalize a series of biomembranes derived from red blood cells, white blood cells, tumor cells, platelets, exosomes and so on. Biomembrane engineering could markedly facilitate the targeted drug delivery, treatment and diagnosis of cancer, inflammation, immunological diseases, bone diseases and Alzheimer's disease. It is anticipated that these membrane modification techniques will advance biomembrane-derived NPs into broader applications in the future.
Collapse
Affiliation(s)
- Ziqing Wu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.,Institute of Medicine, Shanghai University, Shanghai 200444, China.,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Jing Yan
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.,Institute of Medicine, Shanghai University, Shanghai 200444, China.,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai 200444, China.,✉ Corresponding authors: Jiacan Su, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail: ; Yan Wei, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail:
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.,Musculoskeletal Organoid Research Center, Shanghai University, Shanghai 200444, China.,Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.,✉ Corresponding authors: Jiacan Su, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail: ; Yan Wei, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail:
| |
Collapse
|
27
|
Gates BD, Vyletel JB, Zou L, Webber MJ. Multivalent Cucurbituril Dendrons for Cell Membrane Engineering with Supramolecular Receptors. Bioconjug Chem 2022; 33:2262-2268. [PMID: 35802933 PMCID: PMC11144120 DOI: 10.1021/acs.bioconjchem.2c00242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The affinity possible from certain supramolecular motifs rivals that for some of the best-recognized interactions in biology. Cucurbit[7]uril (CB[7]) macrocycles, for example, are capable of achieving affinities in their binding to certain guests that rival that of biotin-avidin. Supramolecular host-guest recognition between CB[7] and certain guests has been demonstrated to spatially localize guest-linked agents to desired sites in vivo, offering opportunities to better exploit this affinity axis for applications in biomedicine. Herein, architectures of CB[7] are prepared from a polyamidoamine (PAMAM) dendrimer scaffold, installing a PEG-linked cholesterol anchor on the opposite end of the dendron to facilitate cell membrane integration. Cells are then modified with this dendritic CB[7] construct in vitro, demonstrating the ability to deliver a model guest-linked agent to the cell membrane. This approach to realize synthetic supramolecular "membrane receptors" may be leveraged in the future for in situ imaging or modulation of cell-based therapies or to facilitate a synthetic supramolecular recognition axis on the cell membrane.
Collapse
Affiliation(s)
- Brant D. Gates
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Jackson B. Vyletel
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Lei Zou
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| | - Matthew J. Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556 USA
| |
Collapse
|
28
|
Burns MWN, Kohler JJ. Engineering Glyco‐Enzymes for Substrate Identification and Targeting. Isr J Chem 2022. [DOI: 10.1002/ijch.202200093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mary W. N. Burns
- Department of Biochemistry UT Southwestern Medical Center Dallas TX 75390 USA
| | - Jennifer J. Kohler
- Department of Biochemistry UT Southwestern Medical Center Dallas TX 75390 USA
| |
Collapse
|
29
|
Chinoy ZS, Friscourt F. Expanding the Strain‐Promoted 1,3‐Dipolar Cycloaddition Arsenal for a More Selective Bioorthogonal Labeling in Living Cells. Isr J Chem 2022. [DOI: 10.1002/ijch.202200055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zoeisha S. Chinoy
- Institut Européen de Chimie et Biologie Université de Bordeaux 2 rue Robert Escarpit 33607 Pessac France
- Institut des Sciences Moléculaires CNRS UMR5255 33405 Talence France
| | - Frédéric Friscourt
- Institut Européen de Chimie et Biologie Université de Bordeaux 2 rue Robert Escarpit 33607 Pessac France
- Institut des Sciences Moléculaires CNRS UMR5255 33405 Talence France
| |
Collapse
|
30
|
Yi W, Xiao P, Liu X, Zhao Z, Sun X, Wang J, Zhou L, Wang G, Cao H, Wang D, Li Y. Recent advances in developing active targeting and multi-functional drug delivery systems via bioorthogonal chemistry. Signal Transduct Target Ther 2022; 7:386. [PMID: 36460660 PMCID: PMC9716178 DOI: 10.1038/s41392-022-01250-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Bioorthogonal chemistry reactions occur in physiological conditions without interfering with normal physiological processes. Through metabolic engineering, bioorthogonal groups can be tagged onto cell membranes, which selectively attach to cargos with paired groups via bioorthogonal reactions. Due to its simplicity, high efficiency, and specificity, bioorthogonal chemistry has demonstrated great application potential in drug delivery. On the one hand, bioorthogonal reactions improve therapeutic agent delivery to target sites, overcoming off-target distribution. On the other hand, nanoparticles and biomolecules can be linked to cell membranes by bioorthogonal reactions, providing approaches to developing multi-functional drug delivery systems (DDSs). In this review, we first describe the principle of labeling cells or pathogenic microorganisms with bioorthogonal groups. We then highlight recent breakthroughs in developing active targeting DDSs to tumors, immune systems, or bacteria by bioorthogonal chemistry, as well as applications of bioorthogonal chemistry in developing functional bio-inspired DDSs (biomimetic DDSs, cell-based DDSs, bacteria-based and phage-based DDSs) and hydrogels. Finally, we discuss the difficulties and prospective direction of bioorthogonal chemistry in drug delivery. We expect this review will help us understand the latest advances in the development of active targeting and multi-functional DDSs using bioorthogonal chemistry and inspire innovative applications of bioorthogonal chemistry in developing smart DDSs for disease treatment.
Collapse
Affiliation(s)
- Wenzhe Yi
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Ping Xiao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Xiaochen Liu
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Zitong Zhao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Xiangshi Sun
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jue Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Lei Zhou
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Guanru Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Haiqiang Cao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Dangge Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000 China
| | - Yaping Li
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264000 China
| |
Collapse
|
31
|
Liu G, Hao M, Zeng B, Liu M, Wang J, Sun S, Liu C, Huilian C. Sialic acid and food allergies: The link between nutrition and immunology. Crit Rev Food Sci Nutr 2022; 64:3880-3906. [PMID: 36369942 DOI: 10.1080/10408398.2022.2136620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Food allergies (FA), a major public health problem recognized by the World Health Organization, affect an estimated 3%-10% of adults and 8% of children worldwide. However, effective treatments for FA are still lacking. Recent advances in glycoimmunology have demonstrated the great potential of sialic acids (SAs) in the treatment of FA. SAs are a group of nine-carbon α-ketoacids usually linked to glycoproteins and glycolipids as terminal glycans. They play an essential role in modulating immune responses and may be an effective target for FA intervention. As exogenous food components, sialylated polysaccharides have anti-FA effects. In contrast, as endogenous components, SAs on immunoglobulin E and immune cell surfaces contribute to the pathogenesis of FA. Given the lack of comprehensive information on the effects of SAs on FA, we reviewed the roles of endogenous and exogenous SAs in the pathogenesis and treatment of FA. In addition, we considered the structure-function relationship of SAs to provide a theoretical basis for the development of SA-based FA treatments.
Collapse
Affiliation(s)
- Guirong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Mengzhen Hao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Binghui Zeng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Manman Liu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Junjuan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shanfeng Sun
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, College of Health and Human Services, San Diego State University, California, United States of America
| | - Che Huilian
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
32
|
Xu J, Zhang Y, Liu Y, You Y, Li F, Chen Y, Xie L, Tong S, Zhou S, Liang K, Huang Y, Jiang G, Song Q, Mei N, Ma F, Gao X, Wang H, Chen J. Vitality-Enhanced Dual-Modal Tracking System Reveals the Dynamic Fate of Mesenchymal Stem Cells for Stroke Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203431. [PMID: 36180405 DOI: 10.1002/smll.202203431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/17/2022] [Indexed: 06/16/2023]
Abstract
Mesenchymal stem cell (MSC) therapy via intravenous transplantation exhibits great potential for brain tissue regeneration, but still faces thorny clinical translation challenges as the unknown dynamic fate leads to the contentious therapeutic mechanism and the poor MSC viability in harsh lesions limits therapeutic efficiency. Here, a vitality-enhanced dual-modal tracking system is designed to improve engraftment efficiency and is utilized to noninvasively explore the fate of intravenous transplanted human umbilical cord-derived MSCs during long-term treatment of ischemic stroke. Such a system is obtained by bioorthogonally conjugating magnetic resonance imaging (MRI) contrast and near-infrared fluorescence (NIRF) imaging nanoparticles to metabolic glycoengineered MSCs with a lipoic acid-containing extracellular antioxidative protective layer. The dynamic fates of MSCs in multi-dimensional space-time evolution are digitally detailed for up to 28 days using MRI and NIRF imaging equipment, and the protective layer greatly shields MSCs from reactive oxygen spices (ROS) degradation, enhances MSC survival, and engraftment efficiency. Additionally, it is observed that the bioengineered MSCs exhibit dynamic intelligent responses corresponding to microenvironment remodeling and exert enhanced therapeutic effects. This dual-modal tracking system enables long-term tracking of MSCs while improving their viability at the lesion sites, which may serve as a valuable tool for expediting the clinical translation of MSC therapy.
Collapse
Affiliation(s)
- Jianpei Xu
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence & Department of Neurology, Zhongshan Hospital, Fudan University, 825 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yipu Liu
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yang You
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Fengan Li
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yu Chen
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Laozhi Xie
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Shiqiang Tong
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Songlei Zhou
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Kaifan Liang
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yukun Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Ni Mei
- Shanghai Center for Drug Evaluation and Inspection, 781 Cailun Road, Shanghai, 201203, P. R. China
| | - Fenfen Ma
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence & Department of Neurology, Zhongshan Hospital, Fudan University, 825 Zhangheng Road, Shanghai, 201203, P. R. China
| | - Jun Chen
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, 826 Zhangheng Road, Shanghai, 201203, P. R. China
| |
Collapse
|
33
|
Yazdi MK, Sajadi SM, Seidi F, Rabiee N, Fatahi Y, Rabiee M, Dominic C.D. M, Zarrintaj P, Formela K, Saeb MR, Bencherif SA. Clickable Polysaccharides for Biomedical Applications: A Comprehensive Review. Prog Polym Sci 2022; 133:101590. [PMID: 37779922 PMCID: PMC10540641 DOI: 10.1016/j.progpolymsci.2022.101590] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent advances in materials science and engineering highlight the importance of designing sophisticated biomaterials with well-defined architectures and tunable properties for emerging biomedical applications. Click chemistry, a powerful method allowing specific and controllable bioorthogonal reactions, has revolutionized our ability to make complex molecular structures with a high level of specificity, selectivity, and yield under mild conditions. These features combined with minimal byproduct formation have enabled the design of a wide range of macromolecular architectures from quick and versatile click reactions. Furthermore, copper-free click chemistry has resulted in a change of paradigm, allowing researchers to perform highly selective chemical reactions in biological environments to further understand the structure and function of cells. In living systems, introducing clickable groups into biomolecules such as polysaccharides (PSA) has been explored as a general approach to conduct medicinal chemistry and potentially help solve healthcare needs. De novo biosynthetic pathways for chemical synthesis have also been exploited and optimized to perform PSA-based bioconjugation inside living cells without interfering with their native processes or functions. This strategy obviates the need for laborious and costly chemical reactions which normally require extensive and time-consuming purification steps. Using these approaches, various PSA-based macromolecules have been manufactured as building blocks for the design of novel biomaterials. Clickable PSA provides a powerful and versatile toolbox for biomaterials scientists and will increasingly play a crucial role in the biomedical field. Specifically, bioclick reactions with PSA have been leveraged for the design of advanced drug delivery systems and minimally invasive injectable hydrogels. In this review article, we have outlined the key aspects and breadth of PSA-derived bioclick reactions as a powerful and versatile toolbox to design advanced polymeric biomaterials for biomedical applications such as molecular imaging, drug delivery, and tissue engineering. Additionally, we have also discussed the past achievements, present developments, and recent trends of clickable PSA-based biomaterials such as 3D printing, as well as their challenges, clinical translatability, and future perspectives.
Collapse
Affiliation(s)
- Mohsen Khodadadi Yazdi
- Jiangsu Co–Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, 210037 Nanjing, China
| | - S. Mohammad Sajadi
- Department of Nutrition, Cihan University-Erbil, Kurdistan Region, 625, Erbil, Iraq
- Department of Phytochemistry, SRC, Soran University, 624, KRG, Iraq
| | - Farzad Seidi
- Jiangsu Co–Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, 210037 Nanjing, China
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rabiee
- Biomaterial group, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Midhun Dominic C.D.
- Department of Chemistry, Sacred Heart College (Autonomous), Kochi, Kerala Pin-682013, India
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, United States
| | - Krzysztof Formela
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
- Sorbonne University, UTC CNRS UMR 7338, Biomechanics and Bioengineering (BMBI), University of Technology of Compiègne, Compiègne, France
| |
Collapse
|
34
|
Murali M, Murali VP, Joseph MM, Rajan S, Maiti KK. Elucidating cell surface glycan imbalance through SERS guided metabolic glycan labelling: An appraisal of metastatic potential in cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112506. [PMID: 35785648 DOI: 10.1016/j.jphotobiol.2022.112506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/14/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
The intrinsic complexities of cell-surface glycans impede tracking the metabolic changes in cells. By coupling metabolic glycan labelling (MGL) and surface-enhanced Raman scattering (SERS), we employed the MGL-SERS strategy to elucidate the differential glycosylation pattern in cancer cell lines. Herein, for the first time, we are reporting an N-alkyl derivative of glucosamine (GlcNPhAlk) as a glycan labelling precursor. The extent of labelling was assessed by utilizing Raman imaging and verified by complementary fluorescence and Western blot analysis. MGL-SERS technique was implemented for a comparative evaluation of cell surface glycan imbalance in different cancer cells wherein a linear relationship between glycan expression and metastatic potential was established. Further, the effect of sialyltransferase inhibitor, P-3Fax-Neu5Ac, on metabolic labelling of GlcNPhAlk proved the incorporation of GlcNPhAlk to the terminal glycans through the sialic acid biosynthetic pathway. Hence, this methodology unveils the phenomenon of metastatic progression in cancer cells with inherent glycosylation-related dysplasia.
Collapse
Affiliation(s)
- Madhukrishnan Murali
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vishnu Priya Murali
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India
| | - Manu M Joseph
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India
| | - Soumya Rajan
- Government College, Kasargod 671123, Kerala, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR- National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram 695019, Kerala, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
35
|
Leong SK, Hsiao JC, Shie JJ. A Multiscale Molecular Dynamic Analysis Reveals the Effect of Sialylation on EGFR Clustering in a CRISPR/Cas9-Derived Model. Int J Mol Sci 2022; 23:ijms23158754. [PMID: 35955894 PMCID: PMC9368999 DOI: 10.3390/ijms23158754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Bacterial and viral pathogens can modulate the glycosylation of key host proteins to facilitate pathogenesis by using various glycosidases, particularly sialidases. Epidermal growth factor receptor (EGFR) signaling is activated by ligand-induced receptor dimerization and oligomerization. Ligand binding induces conformational changes in EGFR, leading to clusters and aggregation. However, information on the relevance of EGFR clustering in the pattern of glycosylation during bacterial and viral invasion remains unclear. In this study, (1) we established CRISPR/Cas9-mediated GFP knock-in (EGFP-KI) HeLa cells expressing fluorescently tagged EGFR at close to endogenous levels to study EGF-induced EGFR clustering and molecular dynamics; (2) We studied the effect of sialylation on EGF-induced EGFR clustering and localization in live cells using a high content analysis platform and raster image correlation spectroscopy (RICS) coupled with a number and brightness (N&B) analysis; (3) Our data reveal that the removal of cell surface sialic acids by sialidase treatment significantly decreases EGF receptor clustering with reduced fluorescence intensity, number, and area of EGFR-GFP clusters per cell upon EGF stimulation. Sialylation appears to mediate EGF-induced EGFR clustering as demonstrated by the change of EGFR-GFP clusters in the diffusion coefficient and molecular brightness, providing new insights into the role of sialylation in EGF-induced EGFR activation; and (4) We envision that the combination of CRISPR/Cas9-mediated fluorescent tagging of endogenous proteins and fluorescence imaging techniques can be the method of choice for studying the molecular dynamics and interactions of proteins in live cells.
Collapse
Affiliation(s)
- Shwee Khuan Leong
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Taiwan International Graduate Program (TIGP), Sustainable Chemical Science & Technology (SCST), Academia Sinica, Taipei 11529, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University (NYCU), Hsinchu 30050, Taiwan
| | - Jye-Chian Hsiao
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Jiun-Jie Shie
- Institute of Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Correspondence:
| |
Collapse
|
36
|
Savelyeva NY, Shpirt AM, Orlova AV, Chizhov AO, Kononov LO. Synthesis of triazole-linked pseudo-oligosialic acid derivatives. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
37
|
Idiago-López J, Moreno-Antolín E, Eceiza M, Aizpurua JM, Grazú V, de la Fuente JM, Fratila RM. From Bench to Cell: A Roadmap for Assessing the Bioorthogonal "Click" Reactivity of Magnetic Nanoparticles for Cell Surface Engineering. Bioconjug Chem 2022; 33:1620-1633. [PMID: 35857350 PMCID: PMC9501912 DOI: 10.1021/acs.bioconjchem.2c00230] [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] [Indexed: 11/28/2022]
Abstract
In this work, we report the use of bioorthogonal chemistry, specifically the strain-promoted click azide-alkyne cycloaddition (SPAAC) for the covalent attachment of magnetic nanoparticles (MNPs) on living cell membranes. Four types of MNPs were prepared, functionalized with two different stabilizing/passivation agents (a polyethylene glycol derivative and a glucopyranoside derivative, respectively) and two types of strained alkynes with different reactivities: a cyclooctyne (CO) derivative and a dibenzocyclooctyne (DBCO) derivative. The MNPs were extensively characterized in terms of physicochemical characteristics, colloidal stability, and click reactivity in suspension. Then, the reactivity of the MNPs toward azide-modified surfaces was evaluated as a closer approach to their final application in a living cell scenario. Finally, the DBCO-modified MNPs, showing superior reactivity in suspension and on surfaces, were selected for cell membrane immobilization via the SPAAC reaction on the membranes of cells engineered to express azide artificial reporters. Overall, our work provides useful insights into the appropriate surface engineering of nanoparticles to ensure a high performance in terms of bioorthogonal reactivity for biological applications.
Collapse
Affiliation(s)
- Javier Idiago-López
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Eduardo Moreno-Antolín
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Maite Eceiza
- Universidad del País Vasco, UPV-EHU, Jose Mari Korta R&D Center, 20018 Donostia San Sebastián, Spain
| | - Jesús M Aizpurua
- Universidad del País Vasco, UPV-EHU, Jose Mari Korta R&D Center, 20018 Donostia San Sebastián, Spain
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Jesús M de la Fuente
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Raluca M Fratila
- Instituto de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/ Pedro Cerbuna 12, 50009 Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 50018 Zaragoza, Spain.,Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| |
Collapse
|
38
|
Antillon K, Ross PA, Farrell MP. Directing CAR NK Cells via the Metabolic Incorporation of CAR Ligands into Malignant Cell Glycans. ACS Chem Biol 2022; 17:1505-1512. [PMID: 35648806 PMCID: PMC10061155 DOI: 10.1021/acschembio.2c00173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The abundance of sialic acid-containing glycans in the glycocalyx of malignant cells enables immune evasion. Here, we leverage the biosynthetic pathways that permit pervasive sialylation to incorporate a chimeric antigen receptor (CAR) ligand into malignant cell glycans, and demonstrate that this increases the susceptibility of malignant cells to the cytolytic activity of CAR-expressing natural killer (NK) cells. Specifically, we applied a C-9-functionalized nonnatural sialic acid [i.e., fluorescein sialic acid (FL-SA)] to modify malignant cell glycans. We confirm the metabolic incorporation of FL-SA into plasma membrane-associated glycans. The preparation of anti-fluorescein CAR NK cells permitted studies demonstrating that treating malignant cells with FL-SA increased susceptibility to CAR NK cell-mediated cytolysis. Furthermore, we observed that the specificity of the anti-fluorescein CAR NK cells is enhanced for fluorescein-labeled cells, and an increased release of cytokines from the CAR NK cells upon incubation with FL-SA-treated cells. The results arising from this study demonstrate that CAR ligands can be metabolically incorporated into malignant cells, and we reason that such strategies could be leveraged to tackle the issue of antigen heterogeneity that limits the clinical efficacy of CAR T/NK cell therapies.
Collapse
Affiliation(s)
- Kathia Antillon
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Patrick A Ross
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mark P Farrell
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| |
Collapse
|
39
|
Comprehensive Analysis of Oligo/Polysialylglycoconjugates in Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23105569. [PMID: 35628382 PMCID: PMC9147586 DOI: 10.3390/ijms23105569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
In cancer cells, cell-surface sialylation is altered, including a change in oligo/polysialic acid (oligo/polySia) structures. Since they are unique and rarely expressed in normal cells, oligo/polySia structures may serve as promising novel biomarkers and targets for therapies. For the diagnosis and treatment of the disease, a precise understanding of the oligo/polySia structures in cancer cells is necessary. In this study, flow cytometric analysis and gene expression datasets were obtained from sixteen different cancer cell lines. These datasets demonstrated the ability to predict glycan structures and their sialylation status. Our results also revealed that sialylation patterns are unique to each cancer cell line. Thus, we can suggest promising combinations of antibody and cancer cell for glycan prediction. However, the precise prediction of minor glycans need to be further explored.
Collapse
|
40
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
41
|
Ying L, Liang C, Zhang Y, Wang J, Wang C, Xia K, Shi K, Yu C, Yang B, Xu H, Zhang Y, Shu J, Huang X, Xing H, Li F, Zhou X, Chen Q. Enhancement of nucleus pulposus repair by glycoengineered adipose-derived mesenchymal cells. Biomaterials 2022; 283:121463. [DOI: 10.1016/j.biomaterials.2022.121463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/28/2022]
|
42
|
Du J, Liu X, Yarema KJ, Jia X. Glycoengineering human neural stem cells (hNSCs) for adhesion improvement using a novel thiol-modified N-acetylmannosamine (ManNAc) analog. BIOMATERIALS ADVANCES 2022; 134:112675. [PMID: 35599100 PMCID: PMC9300770 DOI: 10.1016/j.msec.2022.112675] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
This study sets the stage for the therapeutic use of Ac5ManNTProp, an N-acetylmannosamine (ManNAc) analog that installs thiol-modified sialoglycans onto the surfaces of human neural stem cells (hNSC). First, we compared hNSC adhesion to the extracellular matrix (ECM) proteins laminin, fibronectin, and collagen and found preferential adhesion and concomitant changes to cell morphology and cell spreading for Ac5ManNTProp-treated cells to laminin, compared to fibronectin where there was a modest response, and collagen where there was no observable increase. PCR array transcript analysis identified several classes of cell adhesion molecules that responded to combined Ac5ManNTProp treatment and hNSC adhesion to laminin. Of these, we focused on integrin α6β1 expression, which was most strongly upregulated in analog-treated cells incubated on laminin. We also characterized downstream responses including vinculin display as well as the phosphorylation of focal adhesion kinase (FAK) and extracellular signal-related kinase (ERK). In these experiments, Ac5ManNTProp more strongly induced all tested biological endpoints compared to Ac5ManNTGc, showing that the single methylene unit that structurally separates the two analogs finely tunes biological responses. Together, the concerted modulation of multiple pro-regenerative activities through Ac5ManNTProp treatment, in concert with crosstalk with ECM components, lays a foundation for using our metabolic glycoengineering approach to treat neurological disorders by favorably modulating endpoints that contribute to the viability of transplanted NSCs.
Collapse
Affiliation(s)
- Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Xiao Liu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kevin J. Yarema
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, 21205,Translational Cell and Tissue Engineering Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21231
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
43
|
Jaiswal M, Tran TT, Guo J, Zhou M, Garcia Diaz J, Fanucci GE, Guo Z. Enzymatic glycoengineering-based spin labelling of cell surface sialoglycans to enable their analysis by electron paramagnetic resonance (EPR) spectroscopy. Analyst 2022; 147:784-788. [PMID: 35171149 PMCID: PMC8885856 DOI: 10.1039/d1an02226a] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
A novel method for spin labelling of sialoglycans on the cell surface is described. C9-Azido sialic acid was linked to glycans on live cells via CSTII-catalysed α2,3-sialylation utilizing azido-sialic acid nucleotide as a sialyl donor, which was followed by attachment of a spin label to the azide via click reaction. It enables the study of cell surface sialoglycans by EPR spectroscopy.
Collapse
Affiliation(s)
- Mohit Jaiswal
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Trang T Tran
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Jiatong Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Mingwei Zhou
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Josefina Garcia Diaz
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Gail E Fanucci
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, 214 Leigh Hall, Gainesville, FL 32611, USA.
| |
Collapse
|
44
|
Yu B, Xue X, Yin Z, Cao L, Li M, Huang J. Engineered Cell Membrane-Derived Nanocarriers: The Enhanced Delivery System for Therapeutic Applications. Front Cell Dev Biol 2022; 10:844050. [PMID: 35295856 PMCID: PMC8918578 DOI: 10.3389/fcell.2022.844050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/11/2022] [Indexed: 12/15/2022] Open
Abstract
There has been a rapid development of biomimetic platforms using cell membranes as nanocarriers to camouflage nanoparticles for enhancing bio-interfacial capabilities. Various sources of cell membranes have been explored for natural functions such as circulation and targeting effect. Biomedical applications of cell membranes-based delivery systems are expanding from cancer to multiple diseases. However, the natural properties of cell membranes are still far from achieving desired functions and effects as a nanocarrier platform for various diseases. To obtain multi-functionality and multitasking in complex biological systems, various functionalized modifications of cell membranes are being developed based on physical, chemical, and biological methods. Notably, many research opportunities have been initiated at the interface of multi-technologies and cell membranes, opening a promising frontier in therapeutic applications. Herein, the current exploration of natural cell membrane functionality, the design principles for engineered cell membrane-based delivery systems, and the disease applications are reviewed, with a special focus on the emerging strategies in engineering approaches.
Collapse
Affiliation(s)
- Biao Yu
- The Second Affiliated Hospital, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Xu Xue
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Liehu Cao
- Department of Orthopedics, Luodian Hospital, Shanghai, China
- Department of Orthopedics, Luodian Hospital, Shanghai University, Shanghai, China
| | - Mengmeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jianping Huang
- The Second Affiliated Hospital, Shanghai University, Shanghai, China
- Department of Neurology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
45
|
Ying L, Xu J, Han D, Zhang Q, Hong Z. The Applications of Metabolic Glycoengineering. Front Cell Dev Biol 2022; 10:840831. [PMID: 35252203 PMCID: PMC8892211 DOI: 10.3389/fcell.2022.840831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Mammalian cell membranes are decorated by the glycocalyx, which offer versatile means of generating biochemical signals. By manipulating the set of glycans displayed on cell surface, it is vital for gaining insight into the cellular behavior modulation and medical and biotechnological adhibition. Although genetic engineering is proven to be an effective approach for cell surface modification, the technique is only suitable for natural and genetically encoded molecules. To circumvent these limitations, non-genetic approaches are developed for modifying cell surfaces with unnatural but functional groups. Here, we review latest development of metabolic glycoengineering (MGE), which enriches the chemical functions of the cell surface and is becoming an intriguing new tool for regenerative medicine and tissue engineering. Particular emphasis of this review is placed on discussing current applications and perspectives of MGE.
Collapse
Affiliation(s)
- Liwei Ying
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Junxi Xu
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dawei Han
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Qingguo Zhang
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
- *Correspondence: Qingguo Zhang, ; Zhenghua Hong,
| | - Zhenghua Hong
- Orthopedic Department, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Linhai, China
- *Correspondence: Qingguo Zhang, ; Zhenghua Hong,
| |
Collapse
|
46
|
Au KM, Tisch R, Wang AZ. Immune Checkpoint Ligand Bioengineered Schwann Cells as Antigen-Specific Therapy for Experimental Autoimmune Encephalomyelitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107392. [PMID: 34775659 PMCID: PMC8813901 DOI: 10.1002/adma.202107392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Indexed: 05/05/2023]
Abstract
Failure to establish immune tolerance leads to the development of autoimmune disease. The ability to regulate autoreactive T cells without inducing systemic immunosuppression represents a major challenge in the development of new strategies to treat autoimmune disease. Here, a translational method for bioengineering programmed death-ligand 1 (PD-L1)- and cluster of differentiation 86 (CD86)-functionalized mouse Schwann cells (SCs) to prevent and ameliorate multiple sclerosis (MS) in established mouse models of chronic and relapsing-remitting experimental autoimmune encephalomyelitis (EAE) is described. It is shown that the intravenous (i.v.) administration of immune checkpoint ligand functionalized mouse SCs modifies the course of disease and ameliorates EAE. Further, it is found that such bioengineered mouse SCs inhibit the differentiation of myelin-specific helper T cells into pathogenic T helper type-1 (Th 1) and type-17 (Th 17) cells, promote the development of tolerogenic myelin-specific regulatory T (Treg ) cells, and resolve inflammatory central nervous system microenvironments without inducing systemic immunosuppression.
Collapse
Affiliation(s)
- Kin Man Au
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| | - Roland Tisch
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75230, USA
| |
Collapse
|
47
|
Wang X, Wen C, Davis B, Shi P, Abune L, Lee K, Dong C, Wang Y. Synthetic DNA for Cell Surface Engineering: Experimental Comparison between Click Conjugation and Lipid Insertion in Terms of Cell Viability, Engineering Efficiency, and Displaying Stability. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3900-3909. [PMID: 35020367 DOI: 10.1021/acsami.1c22774] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The cell surface can be engineered with synthetic DNA for various applications ranging from cancer immunotherapy to tissue engineering. However, while elegant methods such as click conjugation and lipid insertion have been developed to engineer the cell surface with DNA, little effort has been made to systematically evaluate and compare these methods. Resultantly, it is often challenging to choose a right method for a certain application or to interpret data from different studies. In this study, we systematically evaluated click conjugation and lipid insertion in terms of cell viability, engineering efficiency, and displaying stability. Cells engineered with both methods can maintain high viability when the concentration of modified DNA is less than 25-50 μM. However, lipid insertion is faster and more efficient in displaying DNA on the cell surface than click conjugation. The efficiency of displaying DNA with lipid insertion is 10-40 times higher than that with click conjugation for a large range of DNA concentration. However, the half-life of physically inserted DNA on the cell surface is 3-4 times lower than that of covalently conjugated DNA, which depends on the working temperature. While the half-life of physically inserted DNA molecules on the cell surface is shorter than that of DNA molecules clicked onto the cell surface, lipid insertion is more effective than click conjugation in the promotion of cell-cell interactions under the two different experimental settings. The data acquired in this work are expected to act as a guideline for choosing an approximate method for engineering the cell surface with synthetic DNA or even other biomolecules.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Brandon Davis
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Peng Shi
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Lidya Abune
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Kyungsene Lee
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| |
Collapse
|
48
|
Wu H, Shajahan A, Yang JY, Capota E, Wands AM, Arthur CM, Stowell SR, Moremen KW, Azadi P, Kohler JJ. A photo-cross-linking GlcNAc analog enables covalent capture of N-linked glycoprotein-binding partners on the cell surface. Cell Chem Biol 2022; 29:84-97.e8. [PMID: 34331854 PMCID: PMC8792112 DOI: 10.1016/j.chembiol.2021.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/11/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
N-glycans are displayed on cell-surface proteins and can engage in direct binding interactions with membrane-bound and secreted glycan-binding proteins (GBPs). Biochemical identification and characterization of glycan-mediated interactions is often made difficult by low binding affinities. Here we describe the metabolic introduction of a diazirine photo-cross-linker onto N-acetylglucosamine (GlcNAc) residues of N-linked glycoproteins on cell surfaces. We characterize sites at which diazirine-modified GlcNAc is incorporated, as well as modest perturbations to glycan structure. We show that diazirine-modified GlcNAc can be used to covalently cross-link two extracellular GBPs, galectin-1 and cholera toxin subunit B, to cell-surface N-linked glycoproteins. The extent of cross-linking correlates with display of the preferred glycan ligands for the GBPs. In addition, covalently cross-linked complexes could be isolated, and protein components of cross-linked N-linked glycoproteins were identified by proteomics analysis. This method may be useful in the discovery and characterization of binding interactions that depend on N-glycans.
Collapse
Affiliation(s)
- Han Wu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA,current affiliation: Agricultural Research Service, U.S. Department of Agriculture, Athens, Georgia 30605
| | - Emanuela Capota
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Amberlyn M. Wands
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Connie M. Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, Harvard Glycomics Center, Harvard Medical School, Boston, MA USA
| | - Sean R. Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, Harvard Glycomics Center, Harvard Medical School, Boston, MA USA
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, Athens, GA 30602 USA
| | - Jennifer J. Kohler
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA,Lead Contact:
| |
Collapse
|
49
|
Wu D, Yang K, Zhang Z, Feng Y, Rao L, Chen X, Yu G. Metal-free bioorthogonal click chemistry in cancer theranostics. Chem Soc Rev 2022; 51:1336-1376. [PMID: 35050284 DOI: 10.1039/d1cs00451d] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bioorthogonal chemistry is a powerful tool to site-specifically activate drugs in living systems. Bioorthogonal reactions between a pair of biologically reactive groups can rapidly and specifically take place in a mild physiological milieu without perturbing inherent biochemical processes. Attributed to their high selectivity and efficiency, bioorthogonal reactions can significantly decrease background signals in bioimaging. Compared with metal-catalyzed bioorthogonal click reactions, metal-free click reactions are more biocompatible without the metal catalyst-induced cytotoxicity. Although a great number of bioorthogonal chemistry-based strategies have been reported for cancer theranostics, a comprehensive review is scarce to highlight the advantages of these strategies. In this review, recent progress in cancer theranostics guided by metal-free bioorthogonal click chemistry will be depicted in detail. The elaborate design as well as the advantages of bioorthogonal chemistry in tumor theranostics are summarized and future prospects in this emerging field are emphasized.
Collapse
Affiliation(s)
- Dan Wu
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou, 310014, P. R. China.
| | - Kuikun Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, P. R. China
| | - Zhankui Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology Hangzhou, 310014, P. R. China.
| | - Yunxuan Feng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, P. R. China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
50
|
He Z, Ishizuka T, Hishikawa Y, Xu Y. Click chemistry for fluorescence imaging via combination of a BODIPY-based ‘turn-on’ probe and a norbornene glucosamine. Chem Commun (Camb) 2022; 58:12479-12482. [DOI: 10.1039/d2cc05359d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present study, we synthesized a novel near-infrared turn-on BODIPY probe and a new norbornene-modified glucosamine derivative.
Collapse
Affiliation(s)
- Zhiyong He
- Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyo-take, Miyazaki 889-1692, Japan
| | - Takumi Ishizuka
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yan Xu
- Medical Sciences, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyo-take, Miyazaki 889-1692, Japan
| |
Collapse
|