1
|
Omotoso MO, Est-Witte SE, Shannon SR, Li S, Nair NM, Neshat SY, Kang SS, Tzeng SY, Green JJ, Schneck JP. Alginate-based artificial antigen presenting cells expand functional CD8 + T cells with memory characteristics for adoptive cell therapy. Biomaterials 2025; 313:122773. [PMID: 39217794 PMCID: PMC11423771 DOI: 10.1016/j.biomaterials.2024.122773] [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: 02/16/2024] [Revised: 07/23/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The development of artificial Antigen Presenting Cells (aAPCs) has led to improvements in adoptive T cell therapy (ACT), an immunotherapy, for cancer treatment. aAPCs help to streamline the consistent production and expansion of T cells, thus reducing the time and costs associated with ACT. However, several issues still exist with ACT, such as insufficient T cell potency, which diminishes the translational potential for ACT. While aAPCs have been used primarily to increase production efficiency of T cells for ACT, the intrinsic properties of a biomaterial-based aAPC may affect T cell phenotype and function. In CD8+ T cells, reactive oxygen species (ROS) and oxidative stress accumulation can activate Forkhead box protein O1 (FOXO1) to transcribe antioxidants which reduce ROS and improve memory formation. Alginate, a biocompatible and antioxidant rich biomaterial, is promising for incorporation into an aAPC formulation to modulate T cell phenotype. To investigate its utility, a novel alginate-based aAPC platform was developed that preferentially expanded CD8+ T cells with memory related features. Alginate-based aAPCs allowed for greater control of CD8+ T cell qualities, including, significantly improved in vivo persistence and augmented in vivo anti-tumor T cell responses.
Collapse
Affiliation(s)
- Mary O Omotoso
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA
| | - Savannah E Est-Witte
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Sydney R Shannon
- Department of Biomedical Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Shuyi Li
- Department of Pathology, School of Medicine, USA; Institute for NanoBioTechnology, USA
| | - Nina M Nair
- Department of Biomedical Engineering, Whiting School of Engineering, USA
| | - Sarah Y Neshat
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Si-Sim Kang
- Department of Pathology, School of Medicine, USA
| | - Stephany Y Tzeng
- Translational Tissue Engineering Center, USA; Department of Biomedical Engineering, Whiting School of Engineering, USA; Johns Hopkins Translational ImmunoEngineering Center, USA
| | - Jordan J Green
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA; Johns Hopkins Translational ImmunoEngineering Center, USA.
| | - Jonathan P Schneck
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA; Johns Hopkins Translational ImmunoEngineering Center, USA; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
2
|
Wang C, Zhang X, Li Q, Hou Y, Sun M, Sun J, Lou Z, Han X, Li Y. A review of carbohydrate polymer-synthesized nanoparticles in cancer immunotherapy: Past, present and future perspectives. Int J Biol Macromol 2025; 286:138195. [PMID: 39645110 DOI: 10.1016/j.ijbiomac.2024.138195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Cancer continues to be a leading factor in mortality and tackling it has been made difficult by the development of immune escape. Furthermore, alternative treatments like surgery, chemotherapy, and radiation have been unsuccessful in eradicating cancer. Despite being effective, they have not succeeded in providing a full cancer treatment and exhibit several negative effects. The field of immunotherapy has been improved by utilizing cancer vaccines, immune checkpoint inhibitors (ICIs), and adoptive cell transfer to enhance immune responses to tumors. Nevertheless, cancer cells need to adapt and become immune to immune reactions, leading to the need for innovative treatment methods. Carbohydrate polymers and their nanoparticles have been beneficial in improving cancer immunotherapy by being customizable to specifically target the immune system. These nanoparticles can change the tumor microenvironment and accelerate immunotherapy by affecting immune cells such as T cells and dendritic cells. Incorporating both chemotherapy and phototherapy into nanoparticles can improve immunotherapy. Furthermore, besides controlling immune reactions, carbohydrate polymer nanoparticles can also be used for theranostic purposes, where they are used to image tumor cells and activate the immune system to eradicate cancer.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Xueyao Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Qiaobei Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Yuxin Hou
- Department of Ultrasonic Diagnosis, The Benxi Hospital of China Medical University, Benxi, China
| | - Minglu Sun
- Department of Ultrasonic Diagnosis, The Cancer Hospital of China Medical University, Shenyang, China
| | - Jun Sun
- Department of Intervention, the Fourth Hospital of China Medical University, Shenyang, China
| | - Zhe Lou
- Department of Cardiovascular Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Liu Z, Li YR, Yang Y, Zhu Y, Yuan W, Hoffman T, Wu Y, Zhu E, Zarubova J, Shen J, Nan H, Yeh KW, Hasani-Sadrabadi MM, Zhu Y, Fang Y, Ge X, Li Z, Soto J, Hsiai T, Yang L, Li S. Viscoelastic synthetic antigen-presenting cells for augmenting the potency of cancer therapies. Nat Biomed Eng 2024; 8:1615-1633. [PMID: 39455719 DOI: 10.1038/s41551-024-01272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
The use of synthetic antigen-presenting cells to activate and expand engineered T cells for the treatment of cancers typically results in therapies that are suboptimal in effectiveness and durability. Here we describe a high-throughput microfluidic system for the fabrication of synthetic cells mimicking the viscoelastic and T-cell-activation properties of antigen-presenting cells. Compared with rigid or elastic microspheres, the synthetic viscoelastic T-cell-activating cells (SynVACs) led to substantial enhancements in the expansion of human CD8+ T cells and to the suppression of the formation of regulatory T cells. Notably, activating and expanding chimaeric antigen receptor (CAR) T cells with SynVACs led to a CAR-transduction efficiency of approximately 90% and to substantial increases in T memory stem cells. The engineered CAR T cells eliminated tumour cells in a mouse model of human lymphoma, suppressed tumour growth in mice with human ovarian cancer xenografts, persisted for longer periods and reduced tumour-recurrence risk. Our findings underscore the crucial roles of viscoelasticity in T-cell engineering and highlight the utility of SynVACs in cancer therapy.
Collapse
Affiliation(s)
- Zeyang Liu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan-Ruide Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Youcheng Yang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Weihao Yuan
- Section of Restorative Dentistry, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Enbo Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jun Shen
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Haochen Nan
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kun-Wei Yeh
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Yichen Zhu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinyang Ge
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhizhong Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Cai J, Zhong H, Luo J, Huang X, Xu Q, Li P. Inhalable multi-stimulus sensitive curcumin-alginate nanogels for scavenging reactive oxygen species and anti-inflammatory co-ordination to alleviate acute lung injury. Int J Biol Macromol 2024; 283:137816. [PMID: 39571867 DOI: 10.1016/j.ijbiomac.2024.137816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024]
Abstract
Acute lung injury (ALI) is one of the most common and extremely critical clinical conditions, which progresses with an inflammatory response and overproduction of reactive oxygen species (ROS), leading to oxidative damage to the lungs. Curcumin (Cur) has great potential in treating ALI due to its excellent antioxidant and anti-inflammatory effects. In this study, Cur and alginate were cross-linked by zinc ions and intermolecular hydrogen bonding to form an inhalable aqueous nanogel system to overcome Cur's low solubility and bioavailability. Cur-alginate (ZA-Cur) nanogels exhibited superior antioxidant properties and down-regulated inflammation-associated factors in vitro with controlled-release behavior under multi-stimulus conditions such as temperature, pH, and ions. Meanwhile, the nanogels system could effectively scavenge cellular ROS to repair oxidative stress damage. In a mice model of ALI, tracheal nebulised inhalation of ZA-Cur nanogels down-regulated the expression of inflammation-related genes such as TNF-α, IL-1β, and IL-6, as well as modulated MDA content and CAT activity to attenuate oxidative stress injury, showing promising lung-protective effects. In conclusion, this work developed inhalable ZA-Cur nanogels to decelerate the progression of lesions in ALI by scavenging intracellular ROS and alleviating inflammation simultaneously, which may be a promising strategy for treating ALI.
Collapse
Affiliation(s)
- Jinyun Cai
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Haiyi Zhong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China.
| | - Jianwei Luo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinghai Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Qiuting Xu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Peiyuan Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China.
| |
Collapse
|
5
|
Wang H, Wen J, Ablimit N, Deng K, Wang W, Jiang W. Degradation of Natural Undaria pinnatifida into Unsaturated Guluronic Acid Oligosaccharides by a Single Alginate Lyase. Mar Drugs 2024; 22:453. [PMID: 39452861 PMCID: PMC11509462 DOI: 10.3390/md22100453] [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: 08/26/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Here, we report on a bifunctional alginate lyase (Vnalg7) expressed in Pichia pastoris, which can degrade natural Undaria pinnatifida into unsaturated guluronic acid di- and trisaccharide without pretreatment. The enzyme activity of Vnalg7 (3620.00 U/mL-culture) was 15.81-fold higher than that of the original alg (228.90 U/mL-culture), following engineering modification. The degradation rate reached 52.75%, and reducing sugar reached 30.30 mg/mL after combining Vnalg7 (200.00 U/mL-culture) and 14% (w/v) U. pinnatifida for 6 h. Analysis of the action mode indicated that Vnalg7 could degrade many substrates to produce a variety of unsaturated alginate oligosaccharides (AOSs), and the minimal substrate was tetrasaccharide. Site-directed mutagenesis showed that Glu238, Glu241, Glu312, Arg236, His307, Lys414, and Tyr418 are essential catalytic sites, while Glu334, Glu344, and Asp311 play auxiliary roles. Mechanism analysis revealed the enzymatic degradation pattern of Vnalg7, which mainly recognizes and attacks the third glycosidic linkage from the reducing end of oligosaccharide substrate. Our findings provide a novel alginate lyase tool and a sustainable and commercial production strategy for value-added biomolecules using seaweeds.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Jiang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Yuan Ming Yuan West Road No. 2, Haidian District, Beijing 100193, China; (H.W.); (J.W.); (N.A.); (K.D.); (W.W.)
| |
Collapse
|
6
|
Pritchard MF, Powell LC, Adams JYM, Menzies G, Khan S, Tøndervik A, Sletta H, Aarstad O, Skjåk-Bræk G, McKenna S, Buurma NJ, Farnell DJJ, Rye PD, Hill KE, Thomas DW. Structure-Activity Relationships of Low Molecular Weight Alginate Oligosaccharide Therapy against Pseudomonas aeruginosa. Biomolecules 2023; 13:1366. [PMID: 37759766 PMCID: PMC10527064 DOI: 10.3390/biom13091366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/26/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Low molecular weight alginate oligosaccharides have been shown to exhibit anti-microbial activity against a range of multi-drug resistant bacteria, including Pseudomonas aeruginosa. Previous studies suggested that the disruption of calcium (Ca2+)-DNA binding within bacterial biofilms and dysregulation of quorum sensing (QS) were key factors in these observed effects. To further investigate the contribution of Ca2+ binding, G-block (OligoG) and M-block alginate oligosaccharides (OligoM) with comparable average size DPn 19 but contrasting Ca2+ binding properties were prepared. Fourier-transform infrared spectroscopy demonstrated prolonged binding of alginate oligosaccharides to the pseudomonal cell membrane even after hydrodynamic shear treatment. Molecular dynamics simulations and isothermal titration calorimetry revealed that OligoG exhibited stronger interactions with bacterial LPS than OligoM, although this difference was not mirrored by differential reductions in bacterial growth. While confocal laser scanning microscopy showed that both agents demonstrated similar dose-dependent reductions in biofilm formation, OligoG exhibited a stronger QS inhibitory effect and increased potentiation of the antibiotic azithromycin in minimum inhibitory concentration and biofilm assays. This study demonstrates that the anti-microbial effects of alginate oligosaccharides are not purely influenced by Ca2+-dependent processes but also by electrostatic interactions that are common to both G-block and M-block structures.
Collapse
Affiliation(s)
- Manon F. Pritchard
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Lydia C. Powell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
- Microbiology and Infectious Disease Group, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Georgina Menzies
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Saira Khan
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Anne Tøndervik
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Håvard Sletta
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Olav Aarstad
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Stephen McKenna
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Niklaas J. Buurma
- Physical Organic Chemistry Centre, School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK;
| | - Damian J. J. Farnell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Philip D. Rye
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway;
| | - Katja E. Hill
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - David W. Thomas
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| |
Collapse
|
7
|
Structural characteristics of native and chemically sulfated polysaccharides from seaweed and their antimelanoma effects. Carbohydr Polym 2022; 289:119436. [DOI: 10.1016/j.carbpol.2022.119436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022]
|
8
|
Chen YB, Zhang YB, Wang YL, Kaur P, Yang BG, Zhu Y, Ye L, Cui YL. A novel inhalable quercetin-alginate nanogel as a promising therapy for acute lung injury. J Nanobiotechnology 2022; 20:272. [PMID: 35690763 PMCID: PMC9187928 DOI: 10.1186/s12951-022-01452-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/06/2022] [Indexed: 02/07/2023] Open
Abstract
Background Acute lung injury (ALI), a severe health-threatening disease, has a risk of causing chronic pulmonary fibrosis. Informative and powerful evidence suggests that inflammation and oxidative stress play a central role in the pathogenesis of ALI. Quercetin is well recognized for its excellent antioxidant and anti-inflammatory properties, which showed great potential for ALI treatment. However, the application of quercetin is often hindered by its low solubility and bioavailability. Therefore, to overcome these challenges, an inhalable quercetin-alginate nanogel (QU-Nanogel) was fabricated, and by this special “material-drug” structure, the solubility and bioavailability of quercetin were significantly enhanced, which could further increase the activity of quercetin and provide a promising therapy for ALI. Results QU-Nanogel is a novel alginate and quercetin based “material-drug” structural inhalable nanogel, in which quercetin was stabilized by hydrogen bonding to obtain a “co-construct” water-soluble nanogel system, showing antioxidant and anti-inflammatory properties. QU-Nanogel has an even distribution in size of less than 100 nm and good biocompatibility, which shows a stronger protective and antioxidant effect in vitro. Tissue distribution results provided evidence that the QU-Nanogel by ultrasonic aerosol inhalation is a feasible approach to targeted pulmonary drug delivery. Moreover, QU-Nanogel was remarkably reversed ALI rats by relieving oxidative stress damage and acting the down-regulation effects of mRNA and protein expression of inflammation cytokines via ultrasonic aerosol inhalation administration. Conclusions In the ALI rat model, this novel nanogel showed an excellent therapeutic effect by ultrasonic aerosol inhalation administration by protecting and reducing pulmonary inflammation, thereby preventing subsequent pulmonary fibrosis. This work demonstrates that this inhalable QU-Nanogel may function as a promising drug delivery strategy in treating ALI. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01452-3. Quercetin (QU)-Nanogel shows a significant therapeutic effect on acute lung injury. Quercetin as an active substance, was also involved in the nanogel construction. The novel nanogel increase the bioavailability of quercetin. Inhalation of QU-Nanogel allows the drug to reach the lungs directly.
Collapse
Affiliation(s)
- Yi-Bing Chen
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West District of Tuanbo New Town, Jinghai District, Tianjin, 301617, China.,First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, 300381, Tianjin, China
| | - Ya-Bin Zhang
- Shandong Provincial Key Laboratory of Fluorine Chemistry and Chemical Materials, School of Chemistry and Chemical Engineering, University of Jinan, 250022, Jinan, China
| | - Yu-Le Wang
- Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, TEDA, 300457, Tianjin, China.,Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Prabhleen Kaur
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Bo-Guang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West District of Tuanbo New Town, Jinghai District, Tianjin, 301617, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, TEDA, 300457, Tianjin, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China.
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10 Poyanghu Road, West District of Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
9
|
Wang M, Chen L, Lou Z, Yuan X, Pan G, Ren X, Wang P. Cloning and Characterization of a Novel Alginate Lyase from Paenibacillus sp. LJ-23. Mar Drugs 2022; 20:md20010066. [PMID: 35049921 PMCID: PMC8780880 DOI: 10.3390/md20010066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 02/01/2023] Open
Abstract
As a low molecular weight alginate, alginate oligosaccharides (AOS) exhibit improved water solubility, better bioavailability, and comprehensive health benefits. In addition, their biocompatibility, biodegradability, non-toxicity, non-immunogenicity, and gelling capability make them an excellent biomaterial with a dual curative effect when applied in a drug delivery system. In this paper, a novel alginate lyase, Algpt, was cloned and characterized from a marine bacterium, Paenibacillus sp. LJ-23. The purified enzyme was composed of 387 amino acid residues, and had a molecular weight of 42.8 kDa. The optimal pH of Algpt was 7.0 and the optimal temperature was 45 °C. The analysis of the conserved domain and the prediction of the three-dimensional structure indicated that Algpt was a novel alginate lyase. The dominant degradation products of Algpt on alginate were AOS dimer to octamer, depending on the incubation time, which demonstrated that Algpt degraded alginate in an endolytic manner. In addition, Algpt was a salt-independent and thermo-tolerant alginate lyase. Its high stability and wide adaptability endow Algpt with great application potential for the efficient preparation of AOS with different sizes and AOS-based products.
Collapse
|
10
|
A Novel PTP1B Inhibitor-Phosphate of Polymannuronic Acid Ameliorates Insulin Resistance by Regulating IRS-1/Akt Signaling. Int J Mol Sci 2021; 22:ijms222312693. [PMID: 34884501 PMCID: PMC8657924 DOI: 10.3390/ijms222312693] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/21/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a critical negative modulator of insulin signaling and has attracted considerable attention in treating type 2 diabetes mellitus (T2DM). Low-molecular-weight polymannuronic acid phosphate (LPMP) was found to be a selective PTP1B inhibitor with an IC50 of 1.02 ± 0.17 μM. Cellular glucose consumption was significantly elevated in insulin-resistant HepG2 cells after LPMP treatment. LPMP could alleviate oxidative stress and endoplasmic reticulum stress, which are associated with the development of insulin resistance. Western blot and polymerase chain reaction (PCR) analysis demonstrated that LPMP could enhance insulin sensitivity through the PTP1B/IRS/Akt transduction pathway. Furthermore, animal study confirmed that LPMP could decrease blood glucose, alleviate insulin resistance, and exert hepatoprotective effects in diabetic mice. Taken together, LPMP can effectively inhibit insulin resistance and has high potential as an anti-diabetic drug candidate to be further developed.
Collapse
|
11
|
Wang M, Chen L, Zhang Z. Potential applications of alginate oligosaccharides for biomedicine - A mini review. Carbohydr Polym 2021; 271:118408. [PMID: 34364551 DOI: 10.1016/j.carbpol.2021.118408] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/23/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023]
Abstract
Extensive research on marine algae, especially on their health-promoting properties, has been conducted. Various ingredients with potential biomedical applications have been discovered and extracted from marine algae. Alginate oligosaccharides are low molecular weight alginate polysaccharides present in cell walls of brown algae. They exhibit various health benefits such as anti-inflammatory, anti-microbial, anti-oxidant, anti-tumor and immunomodulation. Their low-toxicity, non-immunogenicity, and biodegradability make them an excellent material in biomedicine. Alginate oligosaccharides can be chemically or biochemically modified to enhance their biological activity and potential in pharmaceutical applications. This paper provides a brief overview on alginate oligosaccharides characteristics, modification patterns and highlights their vital health promoting properties.
Collapse
Affiliation(s)
- Mingpeng Wang
- College of Life Science, Qufu Normal University, Qufu 273100, China
| | - Lei Chen
- College of Life Science, Qufu Normal University, Qufu 273100, China.
| | - Zhaojie Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|