1
|
Li T, Yang C, Zhang L. Novel comprehensive perspective on Amadori compounds: preparation, multiple roles and interaction with other compounds. Crit Rev Food Sci Nutr 2025:1-24. [PMID: 40269616 DOI: 10.1080/10408398.2025.2494059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Amadori compounds are pivotal intermediates in the Maillard reaction. Amadori compounds serve as flavor enhancers, browning precursors and bioactive components, so they are promising versatile food additives. Comprehensive reviews on multiple roles of Amadori compounds are scarce. Furthermore, there is a lack of reviews on green, efficient and commercially prospective preparation techniques of Amadori compounds and their interactions with other components. This paper reviewed preparation, multiple roles and interactions with other components in foods. Spray drying, microwave heating, natural deep eutectic solvents and vacuum dewatering were deemed as green, efficient and commercially prospective preparation techniques for Amadori compounds. Amadori compounds broadened the application field of Maillard reaction-obtained additives compared to final-products, enabling their uses not only in dark-colored foodstuffs but also in light-colored. Peptide-derived Amadori compounds showed greater potency for flavor generation compared to amino acid-derived. Amadori compounds presented eleven physiological activities. Amadori compounds exerted synergistic effect with essential nutrients (lipids, exogenous amino acids and carbohydrates), functional ingredients (polyphenols, carotenoids, glycosides) as well as several drugs. More preparation approaches of Amadori compounds and their synergistic effects with other ingredients await investigation. This review provided comprehensive theoretical guidance for industrial preparation and application of Amadori compounds as versatile additives.
Collapse
Affiliation(s)
- Tingting Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Cheng Yang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Lianfu Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
2
|
Zhang N, Liu Q, Wang D, Wang X, Pan Z, Han B, He G. Multifaceted roles of Galectins: from carbohydrate binding to targeted cancer therapy. Biomark Res 2025; 13:49. [PMID: 40134029 PMCID: PMC11934519 DOI: 10.1186/s40364-025-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Galectins play pivotal roles in cellular recognition and signaling processes by interacting with glycoconjugates. Extensive research has highlighted the significance of Galectins in the context of cancer, aiding in the identification of biomarkers for early detection, personalized therapy, and predicting treatment responses. This review offers a comprehensive overview of the structural characteristics, ligand-binding properties, and interacting proteins of Galectins. We delve into their biological functions and examine their roles across various cancer types. Galectins, characterized by a conserved carbohydrate recognition domain (CRD), are divided into prototype, tandem-repeat, and chimera types based on their structural configurations. Prototype Galectins contain a single CRD, tandem-repeat Galectins contain two distinct CRDs linked by a peptide, and the chimera-type Galectin-3 features a unique structural arrangement. The capacity of Galectins to engage in multivalent interactions allows them to regulate a variety of signaling pathways, thereby affecting cell fate and function. In cancer, Galectins contribute to tumor cell transformation, angiogenesis, immune evasion, and metastasis, making them critical targets for therapeutic intervention. This review discusses the multifaceted roles of Galectins in cancer progression and explores current advancements in the development of Galectin-targeted therapies. We also address the challenges and future directions for integrating Galectin research into clinical practice to enhance cancer treatment outcomes. In brief, understanding the complex functions of Galectins in cancer biology opens new avenues for therapeutic strategies. Continued research on Galectin interactions and their pathological roles is essential for developing effective carbohydrate-based treatments and improving clinical interventions for cancer patients.
Collapse
Affiliation(s)
- Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiao Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Daihan Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiaoyun Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhaoping Pan
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Gu He
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
3
|
Yang C, Li S, Li C, Li J, Shi R, Cao J, Zhang L. Preparation of red jujube powder with high content of Amadori compounds and higher antioxidant activity by controlling the Maillard reaction. J Food Sci 2024; 89:2218-2231. [PMID: 38372196 DOI: 10.1111/1750-3841.16985] [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: 08/10/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024]
Abstract
Amadori compounds (ACs) are stable compounds produced in the early stage of the Maillard reaction (MR) with health benefits such as immunomodulatory, antithrombosis, and tumor-preventive effects. Jujube is a medicinal and edible fruit in China. It is rich in free amino acids and reducing sugar, but traditionally, little attention was paid to the formation of ACs when jujube was processed, neither the influence of ACs on health effects. In this paper, we aimed to increase ACs through controlling the MR during different heating processes of jujube powder with adjusted water content and find the most effective AC that contributed to the antioxidant effects of jujube powder. The optimal dry-heating conditions to produce ACs were as follows: The water activity was 0.294, the heating temperature was 90°C, and the time was 120 min. After processing, the ACs content of jujube powder was 18.55 ± 0.19 mg/g dry weight (DW), which was more than 100 times of those in the unheated jujube powder (0.153 ± 0.003 mg/g DW). Besides, the antioxidant activity of jujube powder after dry-heating process was higher than that of unheated one. As the most abundant AC in the dry-heated jujube powder (12.90 ± 0.75 mg/g DW), N-(1-deoxy-d-fructose-1-yl) proline (Fru-Pro) showed the highest antioxidant activity (62% of equivalent l-ascorbic acid) among 12 ACs in ferric reducing antioxidant power assay. This result provided a method to produce jujube product with high content of ACs and confirmed the positive contribution of Fru-Pro to the antioxidant activity of the jujube powder.
Collapse
Affiliation(s)
- Cheng Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Shuo Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Chenyan Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Jing Li
- Food Quality and Safety Department, School of Management, Sanda University, Shanghai, China
| | - Rundongdong Shi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Jialing Cao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Lianfu Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
4
|
Mossine VV, Mawhinney TP. 1-Amino-1-deoxy-d-fructose ("fructosamine") and its derivatives: An update. Adv Carbohydr Chem Biochem 2023; 83:1-26. [PMID: 37968036 DOI: 10.1016/bs.accb.2023.10.001] [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] [Indexed: 11/17/2023]
Abstract
1-Amino-1-deoxy-d-fructose (fructosamine, FN) derivatives are omnipresent in all living organisms, as a result of non-enzymatic condensation and Amadori rearrangement reactions between free glucose and biogenic amines such as amino acids, polypeptides, or aminophospholipids. Over decades, steady interest in fructosamine was largely sustained by its role as a key intermediate structure in the Maillard reaction that is responsible for the organoleptic and nutritional value of thermally processed foods, and for pathophysiological effects of hyperglycemia in diabetes. New trends in fructosamine research include the discovery and engineering of FN-processing enzymes, development of advanced tools for hyperglycemia monitoring, and evaluation of the therapeutic potential of both fructosamines and FN-recognizing proteins. This article covers developments in the field of fructosamine and its derivatives since 2010 and attempts to ascertain challenges in future research.
Collapse
Affiliation(s)
- Valeri V Mossine
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
| | - Thomas P Mawhinney
- Department of Biochemistry, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
5
|
Cao J, Yang C, Zhang J, Zhang L, Tsao R. Amadori compounds: analysis, composition in food and potential health beneficial functions. Crit Rev Food Sci Nutr 2023; 65:406-428. [PMID: 39722481 DOI: 10.1080/10408398.2023.2274949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Amadori compounds (ACs) are key intermediates of the Maillard reaction, and found in various thermally processed foods. Simultaneous analysis of multiple ACs is challenging due to the complex amino acid and carbohydrate compositions, and the different food matrices. Most studies focus on the effects of ACs on food flavor and related sensory properties, but not their biological functions. However, increasing evidence shows that ACs possess various beneficial effects on human health, thus a comprehensive review on the various biological activities is warranted. In this review, we summarized the composition and content of ACs in different foods, their formation and degradation reactions, and discussed the latest advances in analytical methods of ACs and their biological functions related to human health. Limitations and research gaps were identified and future perspectives on ACs research were proposed. This review points to the needs of systematic and comprehensive in vitro and in vivo studies on human health related biological functions of ACs and their mechanisms of action, particularly the synergistic effects with other food components and drugs, and roles in intestinal health and metabolic syndrome.
Collapse
Affiliation(s)
- Jialing Cao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Cheng Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jian Zhang
- College of Food, Shihezi University, Shihezi, China
| | - Lianfu Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- College of Food, Shihezi University, Shihezi, China
| | - Rong Tsao
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, Canada
| |
Collapse
|
6
|
Keizman D, Frenkel M, Peer A, Rosenbaum E, Sarid D, Leibovitch I, Mano R, Yossepowitch O, Wolf I, Geva R, Margel D, Rouvinov K, Stern A, Dresler H, Kushnir I, Eliaz I. Modified Citrus Pectin Treatment in Non-Metastatic Biochemically Relapsed Prostate Cancer: Long-Term Results of a Prospective Phase II Study. Nutrients 2023; 15:3533. [PMID: 37630724 PMCID: PMC10459199 DOI: 10.3390/nu15163533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The optimal therapy for patients with non-metastatic biochemically relapsed prostate cancer (BRPC-M0) after local therapy is elusive. Thus, the evaluation of new non-toxic compounds in BRPC-M0 patients is warranted. PectaSol®-Modified citrus pectin (P-MCP) is a food supplement categorized as GRAS (Generally Recognized As Safe) by the FDA. It is a competitive inhibitor of the galectin-3 protein, which is involved in cancer pathogenesis. In an early report of the present phase 2 study, P-MCP treatment for 6 months led to prostate-specific antigen doubling time (PSADT) improvement in 75% of patients with BRPC-M0. Herein, we report the second long-term treatment phase of an additional 12 months of P-MCP therapy (4.8 g × 3/day orally) in patients without disease progression after the initial 6 months of therapy. Of the 46 patients that entered the second treatment phase, 7 patients withdrew consent and decided to continue therapy out of pocket, and 39 initiated the second treatment phase. After a total of 18 months of P-MCP treatment, 85% (n = 33) had a durable long-term response, with 62% (n = 24) showing decreased/stable PSA, 90% (n = 35) PSADT improvement, and all with negative scans. No patient had grade 3/4 toxicity. In conclusion, P-MCP may have long-term durable efficacy and is safe in BRPC-M0.
Collapse
Affiliation(s)
- Daniel Keizman
- Department of Oncology, Tel Aviv Sourasky Medical Center, School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Moshe Frenkel
- Department of Oncology, Rambam Medical Center, Haifa 3200003, Israel; (M.F.); (A.P.)
| | - Avivit Peer
- Department of Oncology, Rambam Medical Center, Haifa 3200003, Israel; (M.F.); (A.P.)
| | - Eli Rosenbaum
- Department of Oncology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - David Sarid
- Department of Oncology, Tel Aviv Sourasky Medical Center, School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ilan Leibovitch
- Department of Urology, Meir Medical Center, Kfar Saba 4428164, Israel;
| | - Roy Mano
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv 69978, Israel; (R.M.); (O.Y.)
| | - Ofer Yossepowitch
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv 69978, Israel; (R.M.); (O.Y.)
| | - Ido Wolf
- Department of Oncology, Tel Aviv Sourasky Medical Center, School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ravit Geva
- Department of Oncology, Tel Aviv Sourasky Medical Center, School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - David Margel
- Department of Urology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - Keren Rouvinov
- Department of Oncology, Soroka Medical Center, Beer Sheva 8410501, Israel;
| | - Anat Stern
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA 95403, USA; (A.S.); (I.E.)
| | - Hadas Dresler
- Department of Oncology, Shaare Zedek Medical Center, Jerusalem 9124001, Israel;
| | - Igal Kushnir
- Department of Oncology, Meir Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA 95403, USA; (A.S.); (I.E.)
| |
Collapse
|
7
|
Wu H, Zhang T, Li N, Gao J. Cell membrane-based biomimetic vehicles for effective central nervous system target delivery: Insights and challenges. J Control Release 2023; 360:169-184. [PMID: 37343724 DOI: 10.1016/j.jconrel.2023.06.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Central nervous system (CNS) disorders, including brain tumor, ischemic stroke, Alzheimer's disease, and Parkinson's disease, threaten human health. And the existence of the blood-brain barrier (BBB) hinders the delivery of drugs and the design of drug targeting delivery vehicles. Over the past decades, great interest has been given to cell membrane-based biomimetic vehicles since the rise of targeting drug delivery systems and biomimetic nanotechnology. Cell membranes are regarded as natural multifunction biomaterials, and provide potential for targeting delivery design and modification. Cell membrane-based biomimetic vehicles appear timely with the participation of cell membranes and nanoparticles, and raises new lights for BBB recognition and transport, and effective therapy with its biological multifunction and high biocompatibility. This review summarizes existing challenges in CNS target delivery and recent advances of different kinds of cell membrane-based biomimetic vehicles for effective CNS target delivery, and deliberates the BBB targeting mechanism. It also discusses the challenges and possibility of clinical translation, and presents new insights for development.
Collapse
Affiliation(s)
- Honghui Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Ni Li
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315041, Zhejiang, PR China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China; Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315041, Zhejiang, PR China.
| |
Collapse
|
8
|
González-Ayón MA, Rochin-Galaviz A, Zizumbo-López A, Licea-Claverie A. Poly( N-vinylcaprolactam)-Gold Nanorods-5 Fluorouracil Hydrogels: In the Quest of a Material for Topical Therapies against Melanoma Skin Cancer. Pharmaceutics 2023; 15:pharmaceutics15041097. [PMID: 37111585 PMCID: PMC10145490 DOI: 10.3390/pharmaceutics15041097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
Chemically crosslinked hydrogels based on poly(N-vinylcaprolactam) (PNVCL) were synthetized by a photoinitiated chemical method. A galactose-based monomer, 2-lactobionamidoethyl methacrylate (LAMA), and N-vinylpyrrolidone (NVP) were added with the aim to improve the physical and chemical properties of hydrogels. The effects of both comonomers on the swelling ratio (Q), volume phase transition temperature (VPTT), glass transition temperature (Tg), and Young's moduli by mechanical compression below and above the VPTT were studied. Gold nanorods (GNRDs) and 5-fluorouracil (5FU) were embedded into the hydrogels, to study the drug release profiles with and without the excitation of GNRDs by irradiation in the near-infrared region (NIR). Results showed that the addition of LAMA and NVP increased the hydrogels' hydrophilicity, elasticity, and VPTT. The loading of GNRDs in the hydrogels changed the release rate of 5FU when irradiated intermittently with an NIR laser. The present study reports on the preparation of a hydrogel-based platform of PNVCL-GNRDs-5FU as a potential hybrid anticancer hydrogel for chemo/photothermal therapy that could be applied against skin cancer for topical 5FU delivery.
Collapse
Affiliation(s)
- Mirian A González-Ayón
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico
| | - Alondra Rochin-Galaviz
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico
| | - Arturo Zizumbo-López
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico
| | - Angel Licea-Claverie
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico
| |
Collapse
|
9
|
Laderach DJ, Compagno D. Inhibition of galectins in cancer: Biological challenges for their clinical application. Front Immunol 2023; 13:1104625. [PMID: 36703969 PMCID: PMC9872792 DOI: 10.3389/fimmu.2022.1104625] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Galectins play relevant roles in tumor development, progression and metastasis. Accordingly, galectins are certainly enticing targets for medical intervention in cancer. To date, however, clinical trials based on galectin inhibitors reported inconclusive results. This review summarizes the galectin inhibitors currently being evaluated and discusses some of the biological challenges that need to be addressed to improve these strategies for the benefit of cancer patients.
Collapse
Affiliation(s)
- Diego José Laderach
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina,*Correspondence: Diego José Laderach,
| | - Daniel Compagno
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
10
|
Mondal UK, Barchi JJ. Isolipoic acid-linked gold nanoparticles bearing the thomsen friedenreich tumor-associated carbohydrate antigen: Stability and in vitro studies. Front Chem 2022; 10:1002146. [PMID: 36300019 PMCID: PMC9588967 DOI: 10.3389/fchem.2022.1002146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/20/2022] [Indexed: 11/19/2022] Open
Abstract
We have previously prepared gold nanoparticles (AuNPs) bearing the Thomsen-Friedenreich antigen disaccharide (TFag), a pan-carcinoma, Tumor-Associated Carbohydrate Antigen (TACA), as tools for various assays and biological applications. Conjugation to AuNPs typically involves the use of thiols due to the affinity of sulfur for the gold surface of the nanoparticle. While a use of a single thiol-containing ligand bound to the gold surface is standard practice, several studies have shown that ligands bearing multiple thiols can enhance the strength of the conjugation in a nearly linear fashion. (R)-(+)-α-Lipoic acid (LA), a naturally occurring disulfide-containing organic acid that is used as a cofactor in many enzymatic reactions, has been used as a linker to conjugate various molecules to AuNPs through its branched di-thiol system to enhance nanoparticle stability. We sought to use a similar system to increase nanoparticle stability that was devoid of the chiral center in (R)-(+)-α-lipoic acid. Isolipoic acid, an isomer of LA, where the exocyclic pentanoic acid chain is shifted by one carbon on the dithiolane ring to produce an achiral acid, was thought to act similarly as LA without the risk of any contaminating (L)-(−) isomer. We synthesized AuNPs with ligands of both serine and threonine glycoamino acids bearing the TFag linked to isolipoic acid and examined their stability under various conditions. In addition, these particles were shown to bind to Galectin-3 and inhibit the interaction of Galectin-3 with a protein displaying copies of the TFag. These agents should prove useful in the design of potential antimetastatic therapeutics that would benefit from achiral linkers that are geometrically linear and achiral.
Collapse
|
11
|
Multicomponent reaction derived small di- and tri-carbohydrate-based glycomimetics as tools for probing lectin specificity. Glycoconj J 2022; 39:587-597. [PMID: 36001188 DOI: 10.1007/s10719-022-10079-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 08/10/2022] [Indexed: 11/04/2022]
Abstract
Lectins, carbohydrate-binding proteins, play important functions in all forms of life from bacteria and viruses to plants, animals, and humans, participating in cell-cell communication and pathogen binding. In an attempt to modify lectin functions, artificial lectin ligands were made usually as big dendrimeric or cluster multivalent glycomimetic structures. Here we synthesized a novel set of glycomimetic ligands through protection/deprotection multicomponent reactions (MCR) approach. Multivalent di-and tri-carbohydrate glycomimetics containing D-fructose, D-galactose, and D-allose moieties were prepared in 63-96% yield. MCR glycomimetics demonstrated different binding abilities for plant lectins Con A and UEA I, and human galectin-3. Information gained about the influence of molecule structure, multivalency and optical purity on the lectin binding ability can be used in lectin detection and sensitivity measurements to further facilitate understanding of carbohydrate recognition process.
Collapse
|
12
|
Mossine VV, Kelley SP, Mawhinney TP. β-d-Galacto-pyranosyl-(1→4)-2-amino-2-de-oxy-α-d-gluco-pyran-ose hydro-chloride monohydrate (lactosamine). IUCRDATA 2022; 7:x220061. [PMID: 36337090 PMCID: PMC9028551 DOI: 10.1107/s241431462200061x] [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: 11/03/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
The title compound, C12H24NO10 +·Cl-·H2O, (I), crystallizes in the monoclinic space group P21 and exists as a monohydrate of a monosubstituted ammonium chloride salt, with the reducing carbohydrate portion existing exclusively as the α-pyran-ose tautomer. The glycosidic bond geometry in (I) is stabilized by an intra-molecular hydrogen bond and is close to that found in crystalline α-lactose. All heteroatoms except gluco-pyran-ose ring O4 participate in an extensive hydrogen-bonding network, which propagates in all directions in the crystal structure of (I).
Collapse
Affiliation(s)
- Valeri V. Mossine
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Steven P. Kelley
- Department of Chemistry, University of Missouri, Columbia, MO 65211, USA
| | | |
Collapse
|
13
|
Expression and Impact of C1GalT1 in Cancer Development and Progression. Cancers (Basel) 2021; 13:cancers13246305. [PMID: 34944925 PMCID: PMC8699795 DOI: 10.3390/cancers13246305] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary C1GalT1 is one of the enzymes that catalyze the addition of sugar residues to proteins (protein glycosylation). It specifically controls the synthesis and formation of a special disaccharide structure Galβ1,3GalNAcα-, which occurs predominately in cancer but rarely in normal cells. Recent studies have shown that C1GalT1 is overexpressed in many common cancers including colon, breast, gastric, lung, head and neck, pancreatic, esophageal, prostate, and hepatocellular cancer. C1GalT1 overexpression is also often associated with poorer prognosis and poorer patient survival. This review summarizes our current understanding of the expression of C1GalT1 in various cancers and discusses the impact of C1GalT change on cancer cell activities in cancer development and progression. Abstract C1GalT1 (T-synthase) is one of the key glycosyltransferases in the biosynthesis of O-linked mucin-type glycans of glycoproteins. It controls the formation of Core-1 disaccharide Galβ1,3GalNAcα- (Thomsen–Friedenreich oncofetal antigen, T or TF antigen) and Core-1-associated carbohydrate structures. Recent studies have shown that C1GalT1 is overexpressed in many cancers of epithelial origin including colon, breast, gastric, head and neck, pancreatic, esophageal, prostate, and hepatocellular cancer. Overexpression of C1GalT1 is often seen to also be associated with poorer prognosis and poorer patient survival. Change of C1GalT1 expression causes glycosylation changes of many cell membrane glycoproteins including mucin proteins, growth factor receptors, adhesion molecules, and death receptors. This leads to alteration of the interactions of these cell surface molecules with their binding ligands, resulting in changes of cancer cell activity and behaviors. This review summarizes our current understanding of the expression of C1GalT1 in various cancers and discusses the impact of C1GalT change on cancer cell activities in cancer development and progression.
Collapse
|
14
|
Keizman D, Frenkel M, Peer A, Kushnir I, Rosenbaum E, Sarid D, Leibovitch I, Mano R, Yossepowitch O, Margel D, Wolf I, Geva R, Dresler H, Rouvinov K, Rapoport N, Eliaz I. Modified Citrus Pectin Treatment in Non-Metastatic Biochemically Relapsed Prostate Cancer: Results of a Prospective Phase II Study. Nutrients 2021; 13:nu13124295. [PMID: 34959847 PMCID: PMC8706421 DOI: 10.3390/nu13124295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Optimal therapy of biochemically relapsed prostate cancer (BRPC) after local treatment is elusive. An established modified citrus pectin (PectaSol®, P-MCP), a dietary polysaccharide, is an established antagonist of galectin-3, a carbohydrate-binding protein involved in cancer pathogenesis. Based on PSA dynamics, we report on the safety and the primary outcome analysis of a prospective phase II study of P-MCP in non-metastatic BRPC based. Sixty patients were enrolled, and one patient withdrew after a month. Patients (n = 59) were given P-MCP, 4.8 grams X 3/day, for six months. The primary endpoint was the rate without PSA progression and improved PSA doubling time (PSADT). Secondary endpoints were the rate without radiologic progression and toxicity. Patients that did not progress by PSA and radiologically at six months continued for an additional twelve months. After six months, 78% (n = 46) responded to therapy, with a decreased/stable PSA in 58% (n = 34), or improvement of PSADT in 75% (n = 44), and with negative scans, and entered the second twelve months treatment phase. Median PSADT improved significantly (p = 0.003). Disease progression during the first 6 months was noted in only 22% (n = 13), with PSA progression in 17% (n = 10), and PSA and radiologic progression in 5% (n = 3). No patients developed grade 3 or 4 toxicity.
Collapse
Affiliation(s)
- Daniel Keizman
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
- Correspondence:
| | - Moshe Frenkel
- Department of Oncology, Rambam Medical Center, Haifa 3109601, Israel; (M.F.); (A.P.)
| | - Avivit Peer
- Department of Oncology, Rambam Medical Center, Haifa 3109601, Israel; (M.F.); (A.P.)
| | - Igal Kushnir
- Department of Oncology, Meir Medical Center and Sackler School of Medicine, Tel-Aviv University, Kfar-Saba 4428164, Israel; (I.K.); (N.R.)
| | - Eli Rosenbaum
- Department of Oncology, Rabin Medical Center, Petah-Tikva 4941492, Israel;
| | - David Sarid
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ilan Leibovitch
- Department of Urology, Meir Medical Center, Kfar-Saba 4439246, Israel;
| | - Roy Mano
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 69978, Israel; (R.M.); (O.Y.)
| | - Ofer Yossepowitch
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 69978, Israel; (R.M.); (O.Y.)
| | - David Margel
- Department of Urology, Rabin Medical Center, Petah-Tikva 4941492, Israel;
| | - Ido Wolf
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ravit Geva
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Hadas Dresler
- Department of Oncology, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
| | - Keren Rouvinov
- Department of Oncology, Soroka Medical Center, Beer-Sheva 8428760, Israel;
| | - Noa Rapoport
- Department of Oncology, Meir Medical Center and Sackler School of Medicine, Tel-Aviv University, Kfar-Saba 4428164, Israel; (I.K.); (N.R.)
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA 95401, USA;
| |
Collapse
|
15
|
Chen K, Fan Y, Gu J, Han Z, Wang Y, Gao L, Zeng H, Mao C, Wang C. Effect of lgals3a on embryo development of zebrafish. Transgenic Res 2021; 30:739-750. [PMID: 34347236 DOI: 10.1007/s11248-021-00276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Abstract
Our study was aimed to investigate the effects of lgals3a (Gal-3 encoding gene) on the development of zebrafish embryo and its underlying mechanisms. Morpholino (MO) technology was used to inhibit the expression of zebrafish lgals3a, and the effect of lgals3a gene knockdown on zebrafish embryo development and the number of monocyte macrophages was observed. Effect of lgals3a-e3i3-MO on apoptosis of zebrafish was detected by acridine orange staining. In addition, the mRNA expression levels of Wnt/β-catenin signaling pathway-related genes were detected by RT-qPCR. Compared with control-MO group, the zebrafish embryos injected with lgals3a-e3i3-MO had obvious defects in the head, eyes, and tail, and pericardial edema. Lgals3a-e3i3-MO significantly reduced the number of mononuclear macrophages in zebrafish embryos compared with the control-MO group. The results of acridine orange staining showed that compared with the control-MO group, lgals3a-e3i3-MO promoted cardiomyocyte apoptosis in zebrafish. Furthermore, lgals3a-e3i3-MO significantly up-regulated the expression of dkk1b, wnt9a, lrp5, fzd7a, β-catenin, Gsk-3β, mycn, myca in the Wnt/β-catenin pathway, and decreased the expression of lef1. These results indicate that lgals3a-e3i3-MO inhibits zebrafish embryo development, reduces the number of mononuclear macrophages, activates Wnt/β-catenin signaling pathway and promotes cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Kan Chen
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yuqi Fan
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Jun Gu
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Zhihua Han
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Lin Gao
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Huasu Zeng
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chengyu Mao
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Changqian Wang
- Department of Cardiology, Shanghai Ninth People's Hospital Affiliated Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
16
|
Chen L, Hong W, Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct Target Ther 2021; 6:225. [PMID: 34099630 PMCID: PMC8182741 DOI: 10.1038/s41392-021-00631-2] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023] Open
Abstract
Over the past decades, great interest has been given to biomimetic nanoparticles (BNPs) since the rise of targeted drug delivery systems and biomimetic nanotechnology. Biological vectors including cell membranes, extracellular vesicles (EVs), and viruses are considered promising candidates for targeted delivery owing to their biocompatibility and biodegradability. BNPs, the integration of biological vectors and functional agents, are anticipated to load cargos or camouflage synthetic nanoparticles to achieve targeted delivery. Despite their excellent intrinsic properties, natural vectors are deliberately modified to endow multiple functions such as good permeability, improved loading capability, and high specificity. Through structural modification and transformation of the vectors, they are pervasively utilized as more effective vehicles that can deliver contrast agents, chemotherapy drugs, nucleic acids, and genes to target sites for refractory disease therapy. This review summarizes recent advances in targeted delivery vectors based on cell membranes, EVs, and viruses, highlighting the potential applications of BNPs in the fields of biomedical imaging and therapy industry, as well as discussing the possibility of clinical translation and exploitation trend of these BNPs.
Collapse
Affiliation(s)
- Li Chen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiqi Hong
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Ren
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
Kim SJ, Chun KH. Non-classical role of Galectin-3 in cancer progression: translocation to nucleus by carbohydrate-recognition independent manner. BMB Rep 2021. [PMID: 32172730 PMCID: PMC7196190 DOI: 10.5483/bmbrep.2020.53.4.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Galectin-3 is a carbohydrate-binding protein and regulates diverse functions, including cell proliferation and differentiation, mRNA splicing, apoptosis induction, immune surveillance and inflammation, cell adhesion, angiogenesis, and cancer-cell metastasis. Galectin-3 is also recommended as a diagnostic or prognostic biomarker of various diseases, including heart disease, kidney disease, and cancer. Galectin-3 exists as a cytosol, is secreted in extracellular spaces on cells, and is also detected in nuclei. It has been found that galectin-3 has different functions in cellular localization: (i) Extracellular galectin-3 mediates cell attachment and detachment. (ii) cytosolic galectin-3 regulates cell survival by blocking the intrinsic apoptotic pathway, and (iii) nuclear galectin-3 supports the ability of the transcriptional factor for target gene expression. In this review, we focused on the role of galectin-3 on translocation from cytosol to nucleus, because it happens in a way independent of carbohydrate recognition and accelerates cancer progression. We also suggested here that intracellular galecin-3 could be a potent therapeutic target in cancer therapy. [BMB Reports 2020; 53(4): 173-180].
Collapse
Affiliation(s)
- Seok-Jun Kim
- Department of Biomedical Science, College of Natural Science, Chosun University; Department of Life Science & Brain Korea 21 Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju 61452, Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
18
|
Nandi S, Ghosh S, Ranjan A, Sood RS, Pal JK, Hajela K, Gupta RK. Lectins in Health and Diseases: Galectins and Cancer. LECTINS 2021:215-271. [DOI: 10.1007/978-981-16-7462-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Kumar S, Mongia A, Gulati S, Singh P, Diwan A, Shukla S. Emerging theranostic gold nanostructures to combat cancer: Novel probes for Combinatorial Immunotherapy and Photothermal Therapy. Cancer Treat Res Commun 2020; 25:100258. [PMID: 33307507 DOI: 10.1016/j.ctarc.2020.100258] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/16/2020] [Accepted: 11/28/2020] [Indexed: 12/21/2022]
Abstract
The application of gold nanoparticles in immunotherapy has emerged as one of the most effective therapeutic strategy for eradicating cancer by releasing antigens, oligonucleotides, adjuvants, immune-stimulating agents into the body. Gold nanoparticles are found to be a superior choice, for generating attack on oncogenic cells, due to their low toxicity, better target specificity, diagnostic capabilities, and enhanced cellular uptake rate. This review focuses on the efficiency of several functionalized gold nanoparticles of diverse shapes and sizes as delivery vehicles to desired target cells through effective immunotherapy, along with a brief discussion about photothermal therapy.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| | - Ayush Mongia
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| | - Shikha Gulati
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| | - Parinita Singh
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| | - Anchita Diwan
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| | - Shefali Shukla
- Department of Chemistry, Sri Venkateswara College, University of Delhi, Delhi, 110021
| |
Collapse
|
20
|
González-Ayón MA, Licea-Claverie A, Sañudo-Barajas JA. Different Strategies for the Preparation of Galactose-Functionalized Thermo-Responsive Nanogels with Potential as Smart Drug Delivery Systems. Polymers (Basel) 2020; 12:E2150. [PMID: 32967249 PMCID: PMC7569999 DOI: 10.3390/polym12092150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Different synthetic strategies were tested for the incorporation of galactose molecules on thermoresponsive nanogels owing to their affinity for receptors expressed in cancer cells. Three families of galactose-functionalized poly(N-vinylcaprolactam) nanogels were prepared with the aim to control the introduction of galactose-moieties into the core, the core-shell interface and the shell. First and second of the above mentioned, were prepared via surfactant free emulsion polymerization (SFEP) by a free-radical mechanism and the third one, via SFEP/reversible addition-fragmentation chain transfer (RAFT) polymerization. Synthetic recipes for the SFEP/free radical method included besides N-vinylcaprolactam (NVCL), a shell forming poly(ethylene glycol) methyl ether methacrylate (PEGMA), while the galactose (GAL) moiety was introduced via 6-O-acryloyl-1,2,:3,4-bis-O-(1-methyl-ethylidene)-α-D-galactopiranose (6-ABG, protected GAL-monomer): nanogels I, or 2-lactobionamidoethyl methacrylate (LAMA, GAL-monomer): nanogels II. For the SFEP/RAFT methodology poly(2-lactobionamidoethyl methacrylate) as GAL macro-chain transfer agent (PLAMA macro-CTA) was first prepared and on a following stage, the macro-CTA was copolymerized with PEGMA and NVCL, nanogels III. The crosslinker ethylene glycol dimethacrylate (EGDMA) was added in both methodologies for the polymer network construction. Nanogel's sizes obtained resulted between 90 and 370 nm. With higher content of PLAMA macro-CTA or GAL monomer in nanogels, a higher the phase-transition temperature (TVPT) was observed with values ranging from 28 to 46 °C. The ρ-parameter, calculated by the ratio of gyration and hydrodynamic radii from static (SLS) and dynamic (DLS) light scattering measurements, and transmission electron microscopy (TEM) micrographs suggest that core-shell nanogels of flexible chains were obtained; in either spherical (nanogels II and III) or hyperbranched (nanogels I) form.
Collapse
Affiliation(s)
- Mirian A. González-Ayón
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico;
| | - Angel Licea-Claverie
- Centro de Graduados e Investigación en Química, Tecnológico Nacional de México/Instituto Tecnológico de Tijuana, Apartado Postal 1166, Tijuana 22454, Mexico;
| | - J. Adriana Sañudo-Barajas
- Centro de Investigación en Alimentación y Desarrollo, A. C. Carretera a El dorado Km 5.5, Culiacán 80110, Mexico;
| |
Collapse
|
21
|
Galectin-3: an immune checkpoint target for musculoskeletal tumor patients. Cancer Metastasis Rev 2020; 40:297-302. [PMID: 32929561 PMCID: PMC7897198 DOI: 10.1007/s10555-020-09932-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/04/2020] [Indexed: 10/25/2022]
Abstract
In the past decade, the development of immune checkpoint inhibitors in oncological clinical settings was in the forefront. However, the interest in musculoskeletal tumor patients as candidates for checkpoint inhibition remains underserved. Here, we are forwarding evidence proposing that galectin-3 (Gal-3) is an additional immune factor in the checkpoint processes. This review is the result of a large-scale cohort study depicting that overexpression of Gal-3 was widely prevalent in patients with musculoskeletal tumors, whereas T cell infiltrations were generally suppressed in the tumor microenvironment. Targeting Gal-3 would serve as a novel immune checkpoint inhibitor candidate in patients afflicted with aggressive musculoskeletal tumors.
Collapse
|
22
|
Moffett S, Shiao TC, Mousavifar L, Mignani S, Roy R. Aberrant glycosylation patterns on cancer cells: Therapeutic opportunities for glycodendrimers/metallodendrimers oncology. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1659. [PMID: 32776710 DOI: 10.1002/wnan.1659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/25/2020] [Accepted: 06/07/2020] [Indexed: 01/29/2023]
Abstract
Despite exciting discoveries and progresses in drug design against cancer, its cure is still rather elusive and remains one of the humanities major challenges in health care. The safety profiles of common small molecule anti-cancer therapeutics are less than at acceptable levels and limiting deleterious side-effects have to be urgently addressed. This is mainly caused by their incapacity to differentiate healthy cells from cancer cells; hence, the use of high dosage becomes necessary. One possible solution to improve the therapeutic windows of anti-cancer agents undoubtedly resides in modern nanotechnology. This review presents a discussion concerning multivalent carbohydrate-protein interactions as this topic pertains to the fundamental aspects that lead glycoscientists to tackle glyconanoparticles. The second section describes the detailed properties of cancer cells and how their aberrant glycan surfaces differ from those of healthy cells. The third section briefly describes the immune systems, both innate and adaptative, because the numerous displays of cell surface protein receptors necessitate to be addressed from the multivalent angles, a strength full characteristic of nanoparticles. The next chapter presents recent advances in glyconanotechnologies, including glycodendrimers in particular, as they apply to glycobiology and carbohydrate-based cancer vaccines. This was followed by an overview of metallodendrimers and how this rapidly evolving field may contribute to our arsenal of therapeutic tools to fight cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | | | | | | | - René Roy
- Glycovax Pharma Inc, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Galectins in prostate and bladder cancer: tumorigenic roles and clinical opportunities. Nat Rev Urol 2020; 16:433-445. [PMID: 31015643 DOI: 10.1038/s41585-019-0183-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Advanced prostate and bladder cancer are two outstanding unmet medical needs for urological oncologists. The high prevalence of these tumours, lack of effective biomarkers and limited effective treatment options highlight the importance of basic research in these diseases. Galectins are a family of β-galactoside-binding proteins that are frequently altered (upregulated or downregulated) in a wide range of tumours and have roles in different stages of tumour development and progression, including immune evasion. In particular, altered expression levels of different members of the galectin family have been reported in prostate and bladder cancers, which, together with the aberrant glycosylation patterns found in tumour cells and the constituent cell types of the tumour microenvironment, can result in malignant transformation and tumour progression. Understanding the roles of galectin family proteins in the development and progression of prostate and bladder cancer could yield key insights to inform the clinical management of these diseases.
Collapse
|
24
|
Li J, Vasilyeva E, Wiseman SM. Beyond immunohistochemistry and immunocytochemistry: a current perspective on galectin-3 and thyroid cancer. Expert Rev Anticancer Ther 2019; 19:1017-1027. [PMID: 31757172 DOI: 10.1080/14737140.2019.1693270] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Thyroid nodules are very common in the general population, most are benign, and do not require any intervention. However, often a challenge exists in discriminating benign thyroid nodules from cancer, without performing a biopsy or operation. Galectin-3 is a beta-galactoside binding protein that is involved in diverse biological processes and has been found to have increased expression in many human cancer types including thyroid cancer. As a result, recent studies have investigated its utility as a serum biomarker for thyroid cancer, as well as a novel target for in vivo molecular imaging of cancer. Additionally, given its role in tumorigenesis and cancer progression, galectin-3 targeting is currently under investigation for its potential utility as treatment for thyroid cancer.Areas covered: Recent studies of galectin-3 as a serum marker for thyroid cancer diagnosis, and in the preclinical setting as a target for cancer imaging and therapy.Expert opinion: Even though current studies evaluating galectin-3 as a serum marker and target for cancer imaging and therapy are promising, further research is required before it can be adopted into routine clinical use.
Collapse
Affiliation(s)
- Jennifer Li
- Department of Surgery, St. Paul's Hospital & University of British Columbia, Vancouver, Canada
| | - Elizaveta Vasilyeva
- Department of Surgery, St. Paul's Hospital & University of British Columbia, Vancouver, Canada
| | - Sam M Wiseman
- Department of Surgery, St. Paul's Hospital & University of British Columbia, Vancouver, Canada
| |
Collapse
|
25
|
Barton S, Li B, Siuta M, Vaibhav J, Song J, Holt CM, Tomono T, Ukawa M, Kumagai H, Tobita E, Wilson K, Sakuma S, Pham W. SPECIFIC MOLECULAR RECOGNITION AS A STRATEGY TO DELINEATE TUMOR MARGIN USING TOPICALLY APPLIED FLUORESCENCE EMBEDDED NANOPARTICLES. PRECISION NANOMEDICINE 2018; 1:194-207. [PMID: 31773101 DOI: 10.33218/prnano1(3).181009.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The Thomsen-Friedenreich (TF) antigen is a tumor-associated antigen consistently expressed on the apical surface of epithelial-based cancer cells, including pancreatic cancer. In this work, we report the development of multimodal imaging probe, the tripolymer fluorescent nanospheres, whose surface was fabricated with peanut agglutinin (PNA) moieties as TF molecular recognition molecules. Here, we demonstrate that the probe is able to detect TF antigen in human pancreatic cancer tissues and differentiate from normal tissue. What is most noteworthy regarding the probe is its ability to visualize tumor margins defined by epithelial TF antigen expression. Further, in vivo preclinical studies using an orthotopic mouse model of pancreatic cancer suggest the potential use of the nanospheres for laparoscopic imaging of pancreatic cancer tumor margins to enhance surgical resection and improve clinical outcomes.
Collapse
Affiliation(s)
- Shawn Barton
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Bo Li
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Michael Siuta
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Janve Vaibhav
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Jessica Song
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Clinton M Holt
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Takumi Tomono
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan
| | - Masami Ukawa
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan
| | | | - Etsuo Tobita
- Advanced Materials R&D Laboratory, ADEKA Corp., Tokyo, Japan
| | - Kevin Wilson
- Vanderbilt University Institute of Imaging Science, Nashville, TN
| | - Shinji Sakuma
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, Japan
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Nashville, TN.,Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN.,Vanderbilt Ingram Cancer Center, Vanderbilt School of Medicine Nashville, TN.,Vanderbilt Brain Institute, Nashville, Vanderbilt University, TN.,Vanderbilt Institute of Chemical Biology, Nashville, TN
| |
Collapse
|
26
|
Nooshkam M, Babazadeh A, Jooyandeh H. Lactulose: Properties, techno-functional food applications, and food grade delivery system. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.07.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Santarsia S, Grosso AS, Trovão F, Jiménez-Barbero J, Carvalho AL, Nativi C, Marcelo F. Molecular Recognition of a Thomsen-Friedenreich Antigen Mimetic Targeting Human Galectin-3. ChemMedChem 2018; 13:2030-2036. [PMID: 30094951 DOI: 10.1002/cmdc.201800525] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Indexed: 11/11/2022]
Abstract
Overexpression of the Thomsen-Friedenreich (TF) antigen in cell membrane proteins occurs in 90 % of adenocarcinomas. Additionally, the binding of the TF antigen to human galectin-3 (Gal-3), also frequently overexpressed in malignancy, promotes cancer progression and metastasis. In this context, structures that interfere with this specific interaction have the potential to prevent cancer metastasis. A multidisciplinary approach combining the optimized synthesis of a TF antigen mimetic with NMR, X-ray crystallography methods, and isothermal titration calorimetry assays was used to unravel the molecular structural details that govern the Gal-3/TF mimetic interaction. The TF mimetic has a binding affinity for Gal-3 similar to that of the TF natural antigen and retains the binding epitope and bioactive conformation observed for the native antigen. Furthermore, from a thermodynamic perspective, a decrease in the enthalpic contribution was observed for the Gal-3/TF mimetic complex; however, this behavior is compensated by a favorable gain in entropy. From a structural perspective, these results establish our TF mimetic as a scaffold to design multivalent solutions to potentially interfere with Gal-3 aberrant interactions and for likely use in hampering Gal-3-mediated cancer cell adhesion and metastasis.
Collapse
Affiliation(s)
- Sabrina Santarsia
- Department of Chemistry Ugo Schiff, University of Florence, Via della Lastruccia, 13-50019, Sesto Fiorentino, Italy
| | - Ana Sofia Grosso
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Filipa Trovão
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Jesús Jiménez-Barbero
- CIC-bioGUNE Bizkaia, 48160, Derio, Spain.,Ikerbasque, Basque Foundation for Science, 48005, Bilbao, Spain.,Department of Organic Chemistry II, EHU-UPV, 48040, Leioa, Spain
| | - Ana Luísa Carvalho
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Cristina Nativi
- Department of Chemistry Ugo Schiff, University of Florence, Via della Lastruccia, 13-50019, Sesto Fiorentino, Italy
| | - Filipa Marcelo
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade De Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| |
Collapse
|
28
|
Restuccia A, Fettis MM, Farhadi SA, Molinaro MD, Kane B, Hudalla GA. Evaluation of Self-Assembled Glycopeptide Nanofibers Modified with N,N′-Diacetyllactosamine for Selective Galectin-3 Recognition and Inhibition. ACS Biomater Sci Eng 2018. [DOI: 10.1021/acsbiomaterials.8b00611] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Antonietta Restuccia
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Margaret M. Fettis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Shaheen A. Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Matthew D. Molinaro
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Bryant Kane
- Department of Biochemistry and Molecular Biology, University of Florida, 1200 Newell Drive, Gainesville, Florida 32611, United States
| | - Gregory A. Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, PO Box 116131, Biomedical Sciences Building JG56, 1275 Center Drive, Gainesville, Florida 32611, United States
| |
Collapse
|
29
|
Kishor C, Ross RL, Blanchard H. Lactulose as a novel template for anticancer drug development targeting galectins. Chem Biol Drug Des 2018; 92:1801-1808. [PMID: 29888844 DOI: 10.1111/cbdd.13348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/02/2018] [Accepted: 05/19/2018] [Indexed: 01/11/2023]
Abstract
Galectins are carbohydrate binding proteins (lectins), which characteristically bind β-galactosides. Galectins play a role in tumour progression through involvement in proliferation, metastasis, angiogenesis, immune evasion and drug resistance. There is need for inhibitors (antagonists) that are specific for distinct galectins and that can interfere with galectin-carbohydrate interactions during cancer progression. Here, we propose that lactulose, a non-digestible galactose-fructose disaccharide, presents a novel inhibitor scaffold for design of inhibitors against galectins. Thermodynamic evaluation displays binding affinity of lactulose against the galectin-1 and galectin-3 carbohydrate recognition domain (CRD). Crystal structures of galectin-1 and galectin-3 in complex with lactulose reveal for the first time the molecular basis of the galectin-lactulose interactions. Molecular modelling was implemented to propose novel lactulose derivatives as potent anti-cancer agents.
Collapse
Affiliation(s)
- Chandan Kishor
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Renee L Ross
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
30
|
Abstract
Galectin-3 plays a role in tissue inflammation, repair, and fibrosis. This article specifically focuses on heart failure (HF), in which galectin-3 has been shown to be a useful biomarker in prognosis and risk stratification, especially in HF with preserved ejection fraction. Experimental research has shown that galectin-3 directly induces pathologic remodeling of the heart, and is therefore considered a culprit protein in the development of cardiac fibrosis in HF, with potentially relevant clinical implications. In summary, galectin-3 is a biomarker and biotarget in cardiac remodeling and fibrosis and future research will target galectin-3-centered diseases.
Collapse
|
31
|
Nangia-Makker P, Hogan V, Raz A. Galectin-3 and cancer stemness. Glycobiology 2018; 28:172-181. [PMID: 29315388 PMCID: PMC6279147 DOI: 10.1093/glycob/cwy001] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/20/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023] Open
Abstract
Over the last few decades galectin-3, a carbohydrate binding protein, with affinity for N-acetyllactosamine residues, has been unique due to the regulatory roles it performs in processes associated with tumor progression and metastasis such as cell proliferation, homotypic/heterotypic aggregation, dynamic cellular transformation, migration and invasion, survival and apoptosis. Structure-function association of galectin-3 reveals that it consists of a short amino terminal motif, which regulates its nuclear-cytoplasmic shuttling; a collagen α-like domain, susceptible to cleavage by matrix metalloproteases and prostate specific antigen; accountable for its oligomerization and lattice formation, and a carbohydrate-recognition/binding domain containing the anti-death motif of the Bcl2 protein family. This structural complexity permits galectin-3 to associate with numerous molecules utilizing protein-protein and/or protein-carbohydrate interactions in the extra-cellular as well as intracellular milieu and regulate diverse signaling pathways, a number of which appear directed towards epithelial-mesenchymal transition and cancer stemness. Self-renewal, differentiation, long-term culturing and drug-resistance potential characterize cancer stem cells (CSCs), a small cell subpopulation within the tumor that is thought to be accountable for heterogeneity, recurrence and metastasis of tumors. Despite the fact that association of galectin-3 to the tumor stemness phenomenon is still in its infancy, there is sufficient direct evidence of its regulatory roles in CSC-associated phenotypes and signaling pathways. In this review, we have highlighted the available data on galectin-3 regulated functions pertinent to cancer stemness and explored the opportunities of its exploitation as a CSC marker and a therapeutic target.
Collapse
Affiliation(s)
- Pratima Nangia-Makker
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
- Karmanos Cancer Institute, 421 East Canfield, Wayne State University, Detroit, MI, USA
| | - Victor Hogan
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
| | - Avraham Raz
- Department of Oncology, School of Medicine, Wayne State University, Karmanos Cancer Institute, 421 East Canfield, Detroit, MI, USA
- Karmanos Cancer Institute, 421 East Canfield, Wayne State University, Detroit, MI, USA
- Department of Pathology, School of Medicine, 540 East Canfield, Wayne State University, Detroit, MI, USA
| |
Collapse
|
32
|
Karacosta LG, Fisk JC, Jessee J, Tati S, Turner B, Ghazal D, Ludwig R, Johnson H, Adams J, Sajjad M, Koury S, Roy R, Olson JR, Rittenhouse-Olson K. Preclinical Analysis of JAA-F11, a Specific Anti-Thomsen-Friedenreich Antibody via Immunohistochemistry and In Vivo Imaging. Transl Oncol 2018; 11:450-466. [PMID: 29477636 PMCID: PMC5834658 DOI: 10.1016/j.tranon.2018.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 01/20/2023] Open
Abstract
The tumor specificity of JAA-F11, a novel monoclonal antibody specific for the Thomsen-Friedenreich cancer antigen (TF-Ag-alpha linked), has been comprehensively studied by in vitro immunohistochemical (IHC) staining of human tumor and normal tissue microarrays and in vivo biodistribution and imaging by micro-positron emission tomography imaging in breast and lung tumor models in mice. The IHC analysis detailed herein is the comprehensive biological analysis of the tumor specificity of JAA-F11 antibody performed as JAA-F11 is progressing towards preclinical safety testing and clinical trials. Wide tumor reactivity of JAA-F11, relative to the matched mouse IgG3 (control), was observed in 85% of 1269 cases of breast, lung, prostate, colon, bladder, and ovarian cancer. Staining on tissues from breast cancer cases was similar regardless of hormonal or Her2 status, and this is particularly important in finding a target on the currently untargetable triple-negative breast cancer subtype. Humanization of JAA-F11 was recently carried out as explained in a companion paper "Humanization of JAA-F11, a Highly Specific Anti-Thomsen-Friedenreich Pancarcinoma Antibody and In Vitro Efficacy Analysis" (Neoplasia 19: 716-733, 2017), and it was confirmed that humanization did not affect chemical specificity. IHC studies with humanized JAA-F11 showed similar binding to human breast tumor tissues. In vivo imaging and biodistribution studies in a mouse syngeneic breast cancer model and in a mouse-human xenograft lung cancer model with humanized 124I- JAA-F11 construct confirmed in vitro tumor reactivity and specificity. In conclusion, the tumor reactivity of JAA-F11 supports the continued development of JAA-F11 as a targeted cancer therapeutic for multiple cancers, including those with unmet need.
Collapse
Affiliation(s)
| | | | | | | | - Bradley Turner
- Department of Pathology, University of Rochester, Rochester, NY.
| | | | | | | | - Julia Adams
- Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY.
| | - Munawwar Sajjad
- Department of Nuclear Medicine, University at Buffalo, Buffalo, NY.
| | - Steven Koury
- Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY.
| | - Rene Roy
- Glycovax Pharma Inc., Montreal, Quebec, Canada.
| | - James R Olson
- For-Robin, Inc, Buffalo, NY; Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY.
| | - Kate Rittenhouse-Olson
- For-Robin, Inc, Buffalo, NY; Department of Biotechnical and Clinical Laboratory Sciences, University at Buffalo, Buffalo, NY.
| |
Collapse
|
33
|
Gentilini LD, Jaworski FM, Tiraboschi C, Pérez IG, Kotler ML, Chauchereau A, Laderach DJ, Compagno D. Stable and high expression of Galectin-8 tightly controls metastatic progression of prostate cancer. Oncotarget 2018; 8:44654-44668. [PMID: 28591719 PMCID: PMC5546508 DOI: 10.18632/oncotarget.17963] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 04/23/2017] [Indexed: 12/21/2022] Open
Abstract
Two decades ago, Galectin-8 was described as a prostate carcinoma biomarker since it is only expressed in the neoplastic prostate, but not in the healthy tissue. To date, no biological function has been attributed to Galectin-8 that could explain this differential expression. In this study we silenced Galectin-8 in two human prostate cancer cell lines, PC3 and IGR-CaP1, and designed a pre-clinical experimental model that allows monitoring the pathology from its early steps to the long-term metastatic stages. We show for the first time that the natural and conserved expression of Gal-8 in tumour cells is responsible for the metastatic evolution of prostate cancer. In fact, Gal-8 controls the rearrangement of the cytoskeleton and E-Cadherin expression, with a major impact on anoikis and homotypic aggregation of tumour cells, both being essential processes for the survival of circulating tumour cells during metastasis. While localized prostate cancer can be cured, metastatic and advanced disease remains a significant therapeutic challenge, urging for the identification of prognostic markers of the metastatic process. Collectively, our results highlight Galectin-8 as a potential target for anti-metastatic therapy against prostate cancer.
Collapse
Affiliation(s)
- Lucas Daniel Gentilini
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Felipe Martín Jaworski
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Carolina Tiraboschi
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Ignacio González Pérez
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Monica Lidia Kotler
- Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | | | - Diego Jose Laderach
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Daniel Compagno
- Molecular and Functional Glyco-Oncology Laboratory, IQUIBICEN-CONICET, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
34
|
Positive associations between galectin-3 and PSA levels in prostate cancer patients: a prospective clinical study-I. Oncotarget 2018; 7:82266-82272. [PMID: 27741512 PMCID: PMC5347690 DOI: 10.18632/oncotarget.12619] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/20/2016] [Indexed: 01/28/2023] Open
Abstract
Galectin-3 (Gal-3), an oncogenic pro-inflammatory protein, has been suggested as a possible complementary diagnostic candidate to prostate specific antigen (PSA) blood test for prostate cancer patients. The presence of the proteins in the circulation (biomarkers) may elicit an intrinsic humoral immune reaction by generating autoantibodies, which consequently could alter the detection levels. Here, we report the associations of the two prostate cancer biomarkers, Gal-3 and PSA in patients at different clinical states of prostate cancer while taking into account the autoantibody levels. A blind, prospective, single institution, pilot study was conducted. A total of 95 men were classified into 5 groups: healthy controls (Group1), newly diagnosed patients (Group2), no recurrence after local therapy (Group3), rising PSA after local therapy (Group4), and metastatic patients (Group5). Gal-3 and PSA level were divided by their respective autoantibodies, which yielded relative PSA and relative Gal-3 levels. After the adjustments, Spearman's rank correlations and linear regression modeling revealed the positive associations between relative Gal-3 and relative PSA levels among all 95 men combined (rho = 0.446, P < 0.0001; fitted slope 0.448, P < 0.0001), in Group2 (rho = 0.616, P = 0.0050; fitted slope 0.438, P =0.0011), and Group3 (rho = 0.484, P = 0.0360; fitted slope 0.470, P = 0.0187). The data show positive associations of relative Gal-3 and relative PSA levels in prostate cancer patients, notably at early clinical time course. Allowing for the influence of autoantibodies, Gal-3 level might be considered as a potential biomarker since it is positively associated with PSA level.
Collapse
|
35
|
Bose RJ, Paulmurugan R, Moon J, Lee SH, Park H. Cell membrane-coated nanocarriers: the emerging targeted delivery system for cancer theranostics. Drug Discov Today 2018; 23:891-899. [PMID: 29426004 DOI: 10.1016/j.drudis.2018.02.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/06/2018] [Accepted: 02/01/2018] [Indexed: 01/12/2023]
Abstract
Cancer is a leading cause of death worldwide. The use of nanocarriers (NCs) has generated significant interest to improve cancer therapy by targeted delivery. However, conventional NCs in general lack specificity and have poor biodistribution, resulting in low efficacy in cancer therapy. To circumvent this problem, there has been an increasing focus on cancer cell membrane-coated NCs (CCMCNCs), which can deliver therapeutics directly to tumor cells. CCMCNCs comprise active cancer cell surface adhesive molecules combined with other functional proteins, and offer extended blood circulation with robust cell-specific targeting, ensuring enhanced intratumoral penetration and higher tumor-specific accumulation of NCs. In this review, we discuss the preparation, homologous targeting mechanisms, and application of CCMCNCs in targeted cancer therapy.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - James Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Soo-Hong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Owen KL, Parker BS. Beyond the vicious cycle: The role of innate osteoimmunity, automimicry and tumor-inherent changes in dictating bone metastasis. Mol Immunol 2017; 110:57-68. [PMID: 29191489 DOI: 10.1016/j.molimm.2017.11.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022]
Abstract
Bone metastasis is a fatal consequence of a subset of solid malignancies that fail to respond to conventional therapies. While a myriad of factors contribute to osteotropism and disseminated cell survival and outgrowth in bone, efforts to inhibit tumor cell growth in the bone-metastatic niche have largely relied on measures that disrupt the bi-directional interactions between bone resident and tumor cells. However, the targeting of isolated stromal interactions has proven ineffective to date in inhibiting bone-metastatic progression and patient mortality. Osteoimmune regulation is now emerging as a critical determinant of metastatic growth in the bone microenvironment. While this has highlighted the importance of innate immune populations in dictating the temporal development of overt bone metastases, the osteoimmunological processes that underpin tumor cell progression in bone remain severely underexplored. Along with tumor-intrinsic alterations that occur specifically within the bone microenvironment, innate osteoimmunological crosstalk poses an exciting area of future discovery and therapeutic development. Here we review current knowledge of the unique exchange that occurs between bone resident cells, innate immune populations and tumor cells that leads to the establishment of a tumor-permissive milieu.
Collapse
Affiliation(s)
- Katie L Owen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
37
|
Cell surface Thomsen-Friedenreich proteome profiling of metastatic prostate cancer cells reveals potential link with cancer stem cell-like phenotype. Oncotarget 2017; 8:98598-98608. [PMID: 29228713 PMCID: PMC5716753 DOI: 10.18632/oncotarget.21985] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/29/2017] [Indexed: 02/04/2023] Open
Abstract
The tumor-associated Thomsen-Friedenreich glycoantigen (TF-Ag) plays an important role in hematogenous metastasis of multiple cancers. The LTQ Orbitrap LC-MS/MS mass spectrometry analysis of cell surface TF-Ag proteome of metastatic prostate cancer cells reveals that several cell surface glycoproteins expressing this carbohydrate antigen in prostate cancer (CD44, α2 integrin, β1 integrin, CD49f, CD133, CD59, EphA2, CD138, transferrin receptor, profilin) are either known as stem cell markers or control important cancer stem-like cell functions. This outcome points to a potential link between TF-Ag expression and prostate cancer stem-like phenotype. Indeed, selecting prostate cancer cells for TF-Ag expression resulted in the enrichment of cells with stem-like properties such as enhanced clonogenic survival and growth, prostasphere formation under non-differentiating and differentiating conditions, and elevated expression of stem cell markers such as CD44 and CD133. Further, the analysis of the recent literature demonstrates that TF-Ag is a common denominator for multiple prostate cancer stem-like cell populations identified to date and otherwise characterized by distinct molecular signatures. The current paradigm suggests that dissemination of tumor cells with stem-like properties to bone marrow that occurred before surgery and/or radiation therapy is largely responsible for disease recurrence years after radical treatment causing a massive clinical problem in prostate cancer. Thus, developing means for destroying disseminated prostate cancer stem-like cells is an important goal of modern cancer research. The results presented in this study suggest that multiple subpopulation of putative prostate cancer stem-like cells characterized by distinct molecular signatures can be attacked using a single target commonly expressed on these cells, the TF-Ag.
Collapse
|
38
|
Kamili NA, Arthur CM, Gerner-Smidt C, Tafesse E, Blenda A, Dias-Baruffi M, Stowell SR. Key regulators of galectin-glycan interactions. Proteomics 2017; 16:3111-3125. [PMID: 27582340 DOI: 10.1002/pmic.201600116] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 08/15/2016] [Accepted: 08/29/2016] [Indexed: 11/08/2022]
Abstract
Protein-ligand interactions serve as fundamental regulators of numerous biological processes. Among protein-ligand pairs, glycan binding proteins (GBPs) and the glycans they recognize represent unique and highly complex interactions implicated in a broad range of regulatory activities. With few exceptions, cell surface receptors and secreted proteins are heavily glycosylated. As these glycans often represent highly regulatable post-translational modifications, alterations in glycosylation can fundamentally impact GBP recognition. Among GBPs, galectins in particular appear to engage a diverse set of glycan determinants to impact a broad range of biological processes. In this review, we will explore factors that impact galectin activity, including the effect of glycan modification on galectin-glycan interactions.
Collapse
Affiliation(s)
- Nourine A Kamili
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Connie M Arthur
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christian Gerner-Smidt
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eden Tafesse
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna Blenda
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Biology, Erskine College, Due West, SC, USA
| | - Marcelo Dias-Baruffi
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Sean R Stowell
- Center for Transfusion Medicine and Cellular Therapies, Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
39
|
Siddiqui M, Ambre S, Keay SK, Rhyne JM, Zhang CO, Barchi JJ. Glycoamino Acid Analogues of the Thomsen-Friedenreich Tumor-Associated Carbohydrate Antigen: Synthesis and Evaluation of Novel Antiproliferative Factor Glycopeptides. ACS OMEGA 2017; 2:5618-5632. [PMID: 28983523 PMCID: PMC5623948 DOI: 10.1021/acsomega.7b01018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/24/2017] [Indexed: 05/21/2023]
Abstract
Glycoamino acid analogues of the Thomsen-Friedenreich antigen disaccharide, where the 4' and 4″ hydroxyl groups were substituted with fluorine or hydrogen, were synthesized and incorporated into the asialylated antiproliferative factor (as-APF), a biologically active form of APF, a glycopeptide found in the urine of patients with interstitial cystitis. Various strategies were employed to incorporate the fluorine atom at the 4-positions of either the galactose or N-acetylgalactosamine unit of the disaccharide antigen, based on stereochemistry and reactivity. These glycopeptides were evaluated in antiproliferative assays on both primary normal bladder epithelial cells and T24 bladder carcinoma cells. Unlike many previously published substitutions to APF, mono-4'-fluorination of the GalNAc residue did not affect the activity, whereas fluoro-derivatives of the galactose 4″-position or both 4' and 4″ hydroxyls showed a reduced potency relative to the monosubstituted GalNAc derivative. A fourth compound where the 4″ position of galactose was deoxygenated showed a lower potency than the parent and monosubstituted compounds. These results suggest that specific substitutions in the sugar moieties in the APF can be tolerated, and the glycomimetic design of APF analogues can include fluorine in the GalNAc sugar of the disaccharide.
Collapse
Affiliation(s)
- Maqbool
A. Siddiqui
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland 21702, United States
| | - Shailesh Ambre
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland 21702, United States
| | - Susan K. Keay
- Medical
Service, Veterans Administration Maryland Health Care System, Baltimore, Maryland 21201, United States
- Baltimore
Research and Education Foundation, Baltimore, Maryland 21201, United States
- Department of Medicine and Department of
Pathology, University of Maryland School
of Medicine, Baltimore, Maryland 21201, United States
| | - Jeffrey M. Rhyne
- Department of Medicine and Department of
Pathology, University of Maryland School
of Medicine, Baltimore, Maryland 21201, United States
| | - Chen-Ou Zhang
- Department of Medicine and Department of
Pathology, University of Maryland School
of Medicine, Baltimore, Maryland 21201, United States
| | - Joseph J. Barchi
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland 21702, United States
- E-mail: . Phone: 301-846-5905. Fax: 301-846-6033 (J.J.B.)
| |
Collapse
|
40
|
Nakajima K, Kho DH, Yanagawa T, Zimel M, Heath E, Hogan V, Raz A. Galectin-3 in bone tumor microenvironment: a beacon for individual skeletal metastasis management. Cancer Metastasis Rev 2017; 35:333-46. [PMID: 27067726 DOI: 10.1007/s10555-016-9622-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The skeleton is frequently a secondary growth site of disseminated cancers, often leading to painful and devastating clinical outcomes. Metastatic cancer distorts bone marrow homeostasis through tumor-derived factors, which shapes different bone tumor microenvironments depending on the tumor cells' origin. Here, we propose a novel insight on tumor-secreted Galectin-3 (Gal-3) that controls the induction of an inflammatory cascade, differentiation of osteoblasts, osteoclasts, and bone marrow cells, resulting in bone destruction and therapeutic failure. In the approaching era of personalized medicine, the current treatment modalities targeting bone metastatic environments are provided to the patient with limited consideration of the cancer cells' origin. Our new outlook suggests delivering individual tumor microenvironment treatments based on the expression level/activity/functionality of tumor-derived factors, rather than utilizing a commonly shared therapeutic umbrella. The notion of "Gal-3-associated bone remodeling" could be the first step toward a specific personalized therapy for each cancer type generating a different bone niche in patients afflicted with non-curable bone metastasis.
Collapse
Affiliation(s)
- Kosei Nakajima
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
| | - Dong Hyo Kho
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
| | - Takashi Yanagawa
- Department of Orthopaedic Surgery, Graduate School of Medicine, Gunma University, Maebashi, Gunma, 371-8511, Japan
| | - Melissa Zimel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
| | - Elisabeth Heath
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
| | - Victor Hogan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA
| | - Avraham Raz
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA.
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, 48201, USA.
| |
Collapse
|
41
|
Cagnoni AJ, Pérez Sáez JM, Rabinovich GA, Mariño KV. Turning-Off Signaling by Siglecs, Selectins, and Galectins: Chemical Inhibition of Glycan-Dependent Interactions in Cancer. Front Oncol 2016; 6:109. [PMID: 27242953 PMCID: PMC4865499 DOI: 10.3389/fonc.2016.00109] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/18/2016] [Indexed: 12/25/2022] Open
Abstract
Aberrant glycosylation, a common feature associated with malignancy, has been implicated in important events during cancer progression. Our understanding of the role of glycans in cancer has grown exponentially in the last few years, concurrent with important advances in glycomics and glycoproteomic technologies, paving the way for the validation of a number of glycan structures as potential glycobiomarkers. However, the molecular bases underlying cancer-associated glycan modifications are still far from understood. Glycans exhibit a natural heterogeneity, crucial for their diverse functional roles as specific carriers of biologically relevant information. This information is decoded by families of proteins named lectins, including sialic acid-binding immunoglobulin (Ig)-like lectins (siglecs), C-type lectin receptors (CLRs), and galectins. Siglecs are primarily expressed on the surface of immune cells and differentially control innate and adaptive immune responses. Among CLRs, selectins are a family of cell adhesion molecules that mediate interactions between cancer cells and platelets, leukocytes, and endothelial cells, thus facilitating tumor cell invasion and metastasis. Galectins, a family of soluble proteins that bind β-galactoside-containing glycans, have been implicated in diverse events associated with cancer biology such as apoptosis, homotypic cell aggregation, angiogenesis, cell migration, and tumor-immune escape. Consequently, individual members of these lectin families have become promising targets for the design of novel anticancer therapies. During the past decade, a number of inhibitors of lectin–glycan interactions have been developed including small-molecule inhibitors, multivalent saccharide ligands, and more recently peptides and peptidomimetics have offered alternatives for tackling tumor progression. In this article, we review the current status of the discovery and development of chemical lectin inhibitors and discuss novel strategies to limit cancer progression by targeting lectin–glycan interactions.
Collapse
Affiliation(s)
- Alejandro J Cagnoni
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan M Pérez Sáez
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Buenos Aires , Argentina
| |
Collapse
|
42
|
Blanchard H, Bum-Erdene K, Bohari MH, Yu X. Galectin-1 inhibitors and their potential therapeutic applications: a patent review. Expert Opin Ther Pat 2016; 26:537-54. [PMID: 26950805 DOI: 10.1517/13543776.2016.1163338] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Galectins have affinity for β-galactosides. Human galectin-1 is ubiquitously expressed in the body and its expression level can be a marker in disease. Targeted inhibition of galectin-1 gives potential for treatment of inflammatory disorders and anti-cancer therapeutics. AREAS COVERED This review discusses progress in galectin-1 inhibitor discovery and development. Patent applications pertaining to galectin-1 inhibitors are categorised as monovalent- and multivalent-carbohydrate-based inhibitors, peptides- and peptidomimetics. Furthermore, the potential of galectin-1 protein as a therapeutic is discussed along with consideration of the unique challenges that galectin-1 presents, including its monomer-dimer equilibrium and oxidized and reduced forms, with regard to delivering an intact protein to a pathologically relevant site. EXPERT OPINION Significant evidence implicates galectin-1's involvement in cancer progression, inflammation, and host-pathogen interactions. Conserved sequence similarity of the carbohydrate-binding sites of different galectins makes design of specific antagonists (blocking agents/inhibitors of function) difficult. Key challenges pertaining to the therapeutic use of galectin-1 are its monomer-dimer equilibrium, its redox state, and delivery of intact galectin-1 to the desired site. Developing modified forms of galectin-1 has resulted in increased stability and functional potency. Gene and protein therapy approaches that deliver the protein toward the target are under exploration as is exploitation of different inhibitor scaffolds.
Collapse
Affiliation(s)
- Helen Blanchard
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | - Khuchtumur Bum-Erdene
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | | | - Xing Yu
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| |
Collapse
|
43
|
Nakajima K, Kho DH, Yanagawa T, Harazono Y, Hogan V, Chen W, Ali-Fehmi R, Mehra R, Raz A. Galectin-3 Cleavage Alters Bone Remodeling: Different Outcomes in Breast and Prostate Cancer Skeletal Metastasis. Cancer Res 2016; 76:1391-402. [PMID: 26837763 PMCID: PMC4863655 DOI: 10.1158/0008-5472.can-15-1793] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/28/2015] [Indexed: 12/11/2022]
Abstract
Management of bone metastasis remains clinically challenging and requires the identification of new molecular target(s) that can be therapeutically exploited to improve patient outcome. Galectin-3 (Gal-3) has been implicated as a secreted factor that alters the bone microenvironment. Proteolytic cleavage of Gal-3 may also contribute to malignant cellular behaviors, but has not been addressed in cancer metastasis. Here, we report that Gal-3 modulates the osteolytic bone tumor microenvironment in the presence of RANKL. Gal-3 was localized on the osteoclast cell surface, and its suppression by RNAi or a specific antagonist markedly inhibited osteoclast differentiation markers, including tartrate-resistant acid phosphatase, and reduced the number of mature osteoclasts. Structurally, the 158-175 amino acid sequence in the carbohydrate recognition domain (CRD) of Gal-3 was responsible for augmented osteoclastogenesis. During osteoclast maturation, Gal-3 interacted and colocalized with myosin-2A along the surface of cell-cell fusion. Pathologically, bone metastatic cancers expressed and released an intact form of Gal-3, mainly detected in breast cancer bone metastases, as well as a cleaved form, more abundant in prostate cancer bone metastases. Secreted intact Gal-3 interacted with myosin-2A, leading to osteoclastogenesis, whereas a shift to cleaved Gal-3 attenuated the enhancement in osteoclast differentiation. Thus, our studies demonstrate that Gal-3 shapes the bone tumor microenvironment through distinct roles contingent on its cleavage status, and highlight Gal-3 targeting through the CRD as a potential therapeutic strategy for mitigating osteolytic bone remodeling in the metastatic niche.
Collapse
Affiliation(s)
- Kosei Nakajima
- Department of Oncology and Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Dhong Hyo Kho
- Department of Oncology and Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Takashi Yanagawa
- Department of Orthopedic Surgery, Graduate School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Yosuke Harazono
- Maxillofacial Surgery, Department of Maxillofacial Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Victor Hogan
- Department of Oncology and Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Wei Chen
- Biostatistics Core, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Rouba Ali-Fehmi
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Avraham Raz
- Department of Oncology and Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan. Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan.
| |
Collapse
|
44
|
Wang L, Guo XL. Molecular regulation of galectin-3 expression and therapeutic implication in cancer progression. Biomed Pharmacother 2016; 78:165-171. [PMID: 26898438 DOI: 10.1016/j.biopha.2016.01.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/05/2016] [Accepted: 01/13/2016] [Indexed: 12/18/2022] Open
Abstract
Galectin-3, a multifunctional protein, distributes inside and outside cells and plays an important role in tumor cell adhesion, proliferation, differentiation, angiogenesis, and metastasis in multiple tumors. Changes in galectin-3 expression are commonly seen in cancer and pre-cancerous conditions. Therefore, to understand the molecular regulation of galectin-3 expression could aid the development of new approach for cancer treatment. This review summarizes different expression of galectin-3 in cancer cells and patients' serum, the regulation mechanism and the potential therapeutic targets of galectin-3 in cancer progression.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Xiu-Li Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
45
|
Serizawa N, Tian J, Fukada H, Baghy K, Scott F, Chen X, Kiss Z, Olson K, Hsu D, Liu FT, Török NJ, Zhao B, Jiang JX. Galectin 3 regulates HCC cell invasion by RhoA and MLCK activation. J Transl Med 2015; 95:1145-56. [PMID: 26146960 PMCID: PMC4586310 DOI: 10.1038/labinvest.2015.77] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 03/31/2015] [Accepted: 05/13/2015] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) carries a poor prognosis with no effective treatment available other than liver transplantation for selected patients. Vascular invasion of HCC is one of the most important negative predictor of survival. As the regulation of invasion of HCC cells is not well understood, our aim was to study the mechanisms by which galectin 3, a β-galactosidase-binding lectin mediates HCC cell migration. HCC was induced by N-diethylnitrosamine in wild-type and galectin 3(-/-) mice, and tumor formation, histology, and tumor cell invasion were assessed. The galectin 3(-/-) mice developed significantly smaller tumor burden with a less invasive phenotype than the wild-type animals. Galectin 3 was upregulated in the wild-type HCC tumor tissue, but not in the surrounding parenchyma. Galectin 3 expression in HCC was induced by NF-κB transactivation as determined by chromatin immunoprecipitation assays. In vitro studies assessed the pro-migratory effects of galectin 3. The migration of hepatoma cells was significantly decreased after transfection by the galectin 3 siRNA and also after using the Rho kinase inhibitor Y-27632. The reorganization of the actin cytoskeleton, RhoA GTPase activity and the phosphorylation of MLC2 (myosin light chain 2) were decreased in the galectin 3 siRNA-transfected cells. In addition, in vitro and in vivo evidence showed that galectin 3 deficiency reduced hepatoma cell proliferation and increased their apoptosis rate. In conclusion, galectin 3 is an important lectin that is induced in HCC cells, and promotes hepatoma cell motility and invasion by an autocrine pathway. Targeting galectin 3 therefore could be an important novel treatment strategy to halt disease progression.
Collapse
Affiliation(s)
- Nobuko Serizawa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Jijiang Tian
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA,State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hiroo Fukada
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Kornelia Baghy
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Fiona Scott
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Xiangling Chen
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Zsofia Kiss
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Kristin Olson
- Department of Pathology, UC Davis Medical Center, Sacramento, CA
| | - Dan Hsu
- Department of Dermatology, UC Davis Medical Center, Sacramento, CA
| | - Fu-Tong Liu
- Department of Dermatology, UC Davis Medical Center, Sacramento, CA
| | - Natalie J Török
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Joy X. Jiang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, CA
| |
Collapse
|
46
|
Sun W, Li L, Yang Q, Shan W, Zhang Z, Huang Y. G3-C12 Peptide Reverses Galectin-3 from Foe to Friend for Active Targeting Cancer Treatment. Mol Pharm 2015; 12:4124-36. [PMID: 26393405 DOI: 10.1021/acs.molpharmaceut.5b00568] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Galectin-3 is overexpressed by numerous carcinomas and is a potential target for active tumor treatments. On the other hand, galectin-3 also plays a key role in cancer progression and prevents cells from undergoing apoptosis, thereby offsetting the benefits of active targeting drugs. However, the relative contribution of the protective antiapoptotic effects of galectin-3 and the proapoptotic effects of galectin-3-targeted therapies has remained yet unrevealed. Here, we show that a galectin-3-binding peptide G3-C12 could reverse galectin-3 from foe to friend for active targeting delivery system. Results showed G3-C12 modified N-(2-hydroxypropyl)methacrylamide copolymer doxorubicin conjugates (G3-C12-HPMA-Dox) could internalize into galectin-3 overexpressed PC-3 cells via a highly specific ligand-receptor pathway (2.2 times higher cellular internalization than HPMA-Dox). The internalized Dox stimulated the translocation of galectin-3 to the mitochondria to prevent from apoptosis. In turn, this caused G3-C12-HPMA-Dox to concentrate into the mitochondria after binding to galectin-3 intracellularly. Initially, mitochondrial galectin-3 weakened Dox-induced mitochondrial damage; however, as time progressed, G3-C12 active-mediation allowed increasing amounts of Dox to be delivered to the mitochondria, which eventually induced higher level of apoptosis than nontargeted copolymers. In addition, G3-C12 downregulates galectin-3 expression, 0.43 times lower than control cells, which could possibly be responsible for the suppressed cell migration. Thus, G3-C12 peptide exerts sequential targeting to both cell membrane and mitochondria via regulating galectin-3, and eventually reverses and overcomes the protective effects of galectin-3; therefore, it could be a promising agent for the treatment of galectin-3-overexpressing cancers.
Collapse
Affiliation(s)
- Wei Sun
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| | - Lian Li
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| | - Qingqing Yang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| | - Wei Shan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University , No. 17, Block 3, Southern Renmin Road, Chengdu 610041, P. R. China
| |
Collapse
|
47
|
Mori Y, Akita K, Yashiro M, Sawada T, Hirakawa K, Murata T, Nakada H. Binding of Galectin-3, a β-Galactoside-binding Lectin, to MUC1 Protein Enhances Phosphorylation of Extracellular Signal-regulated Kinase 1/2 (ERK1/2) and Akt, Promoting Tumor Cell Malignancy. J Biol Chem 2015; 290:26125-40. [PMID: 26342075 DOI: 10.1074/jbc.m115.651489] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Indexed: 11/06/2022] Open
Abstract
Both mucin 1 (MUC1) and galectin-3 are known to be overexpressed in various malignant tumors and associated with a poor prognosis. It has been extensively reported that MUC1 is involved in potentiation of growth factor-dependent signal transduction. Because some carbohydrate moieties carried on MUC1 change to preferable ones for binding of galectin-3 in cancer cells, we speculated that MUC1-mediated signaling may occur through direct binding of galectin-3. Immunochemical studies showed that the distribution of galectin-3 coincided with that of MUC1 in various human tumor tissues but not in human nonmalignant tissues, and the level of galectin-3 retained on the surface of various cancer cells paralleled that of MUC1. Treatment of MUC1-expressing cells with galectin-3 induced phosphorylation of ERK1/2 and Akt following enhanced phosphorylation of MUC1 C-terminal domain, consistently promoting tumor cell malignancy. It is also noted that this enhanced phosphorylation occurred independently of EGF receptor-mediated signaling in both EGF receptor- and MUC1-expressing cells, and multivalency of galectin-3 was important for initiation of MUC1-mediated signaling. Expectedly, both silencing of endogenous galectin-3 and treatment with galectin-3 antagonists down-regulated cell proliferation of MUC1-expressing cells. These results suggest that the binding of galectin-3 to MUC1 plays a key role in MUC1-mediated signaling. Thus, constitutive activation of MUC1-mediated signaling in an autocrine/paracrine manner caused by ligation of galectin-3 promotes uncontrolled tumor cell malignancy. This signaling may be another MUC1-mediated pathway and function in parallel with a growth factor-dependent MUC1-mediated signaling pathway.
Collapse
Affiliation(s)
- Yugo Mori
- From the Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Kaoru Akita
- From the Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Masakazu Yashiro
- the Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan, the Oncology Institute of Geriatrics and Medical Science, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan, and
| | - Tetsuji Sawada
- the Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Kosei Hirakawa
- the Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Takeomi Murata
- the Department of Applied Biological Chemistry, Faculty of Agriculture, Shizuoka University, Shizuoka 422-8529, Japan
| | - Hiroshi Nakada
- From the Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan,
| |
Collapse
|
48
|
Jayaram S, Kapoor S, Dharmesh SM. Pectic polysaccharide from corn (Zea mays L.) effectively inhibited multi-step mediated cancer cell growth and metastasis. Chem Biol Interact 2015; 235:63-75. [DOI: 10.1016/j.cbi.2015.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 03/23/2015] [Accepted: 04/07/2015] [Indexed: 01/12/2023]
|
49
|
Galectin-3 Overrides PTRF/Cavin-1 Reduction of PC3 Prostate Cancer Cell Migration. PLoS One 2015; 10:e0126056. [PMID: 25942420 PMCID: PMC4420459 DOI: 10.1371/journal.pone.0126056] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/28/2015] [Indexed: 12/31/2022] Open
Abstract
Expression of Caveolin-1 (Cav1), a key component of cell surface caveolae, is elevated in prostate cancer (PCa) and associated with PCa metastasis and a poor prognosis for PCa patients. Polymerase I and Transcript Release Factor (PTRF)/cavin-1 is a cytoplasmic protein required for Cav1-dependent formation of caveolae. Expression of PTRF reduces the motility of PC3 cells, a metastatic prostate cancer cell line that endogenously expresses abundant Cav1 but no PTRF and no caveolae, suggesting a role for non-caveolar Cav1 domains, or Cav1 scaffolds, in PCa cell migration. Tyrosine phosphorylated Cav1 (pCav1) functions in concert with Galectin-3 (Gal3) and the galectin lattice to stabilize focal adhesion kinase (FAK) within focal adhesions (FAs) and promote cancer cell motility. However, whether PTRF regulation of Cav1 function in PCa cell migration is related to Gal3 expression and functionality has yet to be determined. Here we show that PTRF expression in PC3 cells reduces FAK stabilization in focal adhesions and reduces cell motility without affecting pCav1 levels. Exogenous Gal3 stabilized FAK in focal adhesions of PTRF-expressing cells and restored cell motility of PTRF-expressing PC3 cells to levels of PC3 cells in a dose-dependent manner, with an optimal concentration of 2 µg/ml. Exogenous Gal3 stabilized FAK in focal adhesions of Gal3 knockdown PC3 cells but not in Cav1 knockdown PC3 cells. Cav1 knockdown also prevented Gal3 rescue of FA-associated FAK stabilization in PTRF-expressing PC3 cells. Our data support a role for PTRF/cavin-1, through caveolae formation, as an attenuator of the non-caveolar functionality of Cav1 in Gal3-Cav1 signalling and regulation of focal adhesion dynamics and cancer cell migration.
Collapse
|
50
|
Thijssen VL, Heusschen R, Caers J, Griffioen AW. Galectin expression in cancer diagnosis and prognosis: A systematic review. Biochim Biophys Acta Rev Cancer 2015; 1855:235-47. [PMID: 25819524 DOI: 10.1016/j.bbcan.2015.03.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 03/14/2015] [Accepted: 03/16/2015] [Indexed: 02/07/2023]
Abstract
Galectins are a family of proteins that bind to specific glycans thereby deciphering the information captured within the glycome. In the last two decades, several galectin family members have emerged as versatile modulators of tumor progression. This has initiated the development and preclinical assessment of galectin-targeting compounds. With the first compounds now entering clinical trials it is pivotal to gain insight in the diagnostic and prognostic value of galectins in cancer as this will allow a more rational selection of the patients that might benefit most from galectin-targeted therapies. Here, we present a systematic review of galectin expression in human cancer patients. Malignant transformation is frequently associated with altered galectin expression, most notably of galectin-1 and galectin-3. In most cancers, increased galectin-1 expression is associated with poor prognosis while elevated galectin-9 expression is emerging as a marker of favorable disease outcome. The prognostic value of galectin-3 appears to be tumor type dependent and the other galectins require further investigation. Regarding the latter, additional studies using larger patient cohorts are essential to fully unravel the diagnostic and prognostic value of galectin expression. Furthermore, to better compare different findings, consensus should be reached on how to assess galectin expression, not only with regard to localization within the tissue and within cellular compartments but also regarding alternative splicing and genomic variations. Finally, linking galectin expression and function to aberrant glycosylation in cancer cells will improve our understanding of how these versatile proteins can be exploited for diagnostic, prognostic and even therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Victor L Thijssen
- Angiogenesis Laboratory, Department Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands; Angiogenesis Laboratory, Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liege, Liege, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liege, Liege, Belgium
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|