1
|
Ren Y, Mao S, Chen P, Tan W, Ye X, Tian J. Protocatechuic acid/sodium alginate multilayer coating induced by metal ion enhanced the ulcerative colitis alleviations of Lactiplantibacillus plantarum. Int J Biol Macromol 2025; 284:138122. [PMID: 39608540 DOI: 10.1016/j.ijbiomac.2024.138122] [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: 09/18/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Oral intake of probiotics is a promising approach to alleviate colitis. However, environmental sensitivity of the gastrointestinal tract and poor adhesion of probiotics to the intestine hamper the remedial effects. In this study, a simple yet effective novel probiotic multilayer coating consisting of Fe3+-protocatechuic acid (PCA) crosslinked network and Ca2+-induced sodium alginate (SA) for arming Lactiplantibacillus plantarum (LP) was developed. In the dextran sulfate sodium-induced colitis mouse model, SA-PCA-LP effectively alleviated colitis by regulating the expression of inflammatory cytokines, and repairing gut barriers. In addition, SA-PCA-LP regulated the gut microbiota and promoted the production of short-chain fatty acids, which further promoted the remission of colitis. Untargeted metabolomics also revealed that the scymnol, adenosine 5'-monophosphat, guanidylic acid, and 9H-purine-9-ol were significantly up-regulated in SA-PCA-LP group. In general, the novel coating strategies developed in the present study will motivate researchers to arm probiotics with various prebiotics to effectively alleviate colitis.
Collapse
Affiliation(s)
- Yanming Ren
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Shuifang Mao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Pin Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Wen Tan
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Zhejiang University Zhongyuan Institute, Zhengzhou 450000, China.
| | - Jinhu Tian
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Zhejiang University-Wuxi Xishan Modern Agriculture Joint Research Center, Wuxi 214117, China; Zhejiang University-Wuxi Xishan Modern Agriculture Joint Research Center, Wuxi 214117, China.
| |
Collapse
|
2
|
Gu S, Zhao X, Wan F, Gu D, Xie W, Gao C. Intracellularly Gelated Macrophages Loaded with Probiotics for Therapy of Colitis. NANO LETTERS 2024; 24:13504-13512. [PMID: 39418594 DOI: 10.1021/acs.nanolett.4c02699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Probiotics therapy has garnered significant attention in the treatment of inflammatory bowel disease (IBD). However, a large number of oral administrated probiotics are inactivated after passing through the gastric acid environment, and their ability to colonize in the intestine is also weak. Herein, this study develops a novel probiotics formulation (GM-EcN) by incorporating Escherichia coli Nissle 1917 (EcN) into intracellularly gelated macrophages (GM). Intracellular hydrogel is designed to load and prevent EcN from digestion in gastric juice, and GM acts as a macrophage-like carrier to carry the attached probiotics to colonize in the inflammatory intestine. In addition, hydrogel serves as an ideal cytoskeletal structure to maintain the intact cell morphology and membrane structure of GM, comparable to source macrophages. Due to the receptor-ligand interaction, inflammation-related membrane proteins enable GM as a cell sponge to sequestrate and neutralize multiple inflammatory cytokines. In vivo treatment demonstrates that GM-EcN efficiently alleviates IBD symptoms and enhances gut microbiota recovery.
Collapse
Affiliation(s)
- Siyao Gu
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xiaona Zhao
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
- Guangxi University of Chinese Medicine, 530004 Nanning, China
| | - Fang Wan
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Cheng Gao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
| |
Collapse
|
3
|
Luo Z, Cao Y, Liao Z, Gong N, Ma P, Li Z, Lai X, Zhang Y, Zhu X, Li Z, Wu YL, Huo S. Mitochondria-Targeted Gold Biometallization for Photoacoustically Visualized Photothermal Cancer Therapy. ACS NANO 2024; 18:29667-29677. [PMID: 39404617 DOI: 10.1021/acsnano.4c08567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Subcellular biomineralization systems with cellular intervention functions have shown great potential in cancer theranostic applications. However, the lack of subcellular specificity, high ion concentrations, and long incubation time required for biomineralization still limit its in vivo therapeutic efficacy. Herein, we report a mitochondria-targeted polymer-gold complex (TPPM-Au) to realize mitochondrial biometallization, which involves analogous mechanisms during biomineralization, for cancer treatment in vivo. The TPP-containing TPPM-Au delivered more Au3+ selectively into the mitochondria of cancer cells than normal cells, rapidly mineralizing to gold nanoparticles (GNPs) and consuming a large amount of the antioxidant glutathione (GSH). The formed GNPs can further continue consuming GSH with the atomic economy by forming Au-S with GSH, which further results in the accumulation of reactive oxygen species (ROS), thereby impairing mitochondrial function and inducing cell apoptosis. More importantly, TPPM-Au is capable of having superior tumor-penetrating, excellent photothermal and photoacoustic properties, endowing it with the ability to inhibit tumor growth through spatiotemporally monitorable mitochondria-targeted biometallization and photothermal therapy. The mitochondria-targeted gold biometallization theranostic platform provides insight into the application of subcellularly targeted biometallization or biomineralization in cancer therapy.
Collapse
Affiliation(s)
- Zheng Luo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- Engineering (IMRE), A*STAR (Agency for Science, Technology and Research), Institute of Materials Research and Engineering, 138634 Singapore
| | - Yin Cao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zhihuan Liao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Panqin Ma
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zhiguo Li
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xiyu Lai
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Yuhan Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Xuan Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Zibiao Li
- Energy and Environment (ISCE2), A*STAR (Agency for Science, Technology and Research), Institute of Sustainability for Chemicals, 627833 Singapore
- Engineering (IMRE), A*STAR (Agency for Science, Technology and Research), Institute of Materials Research and Engineering, 138634 Singapore
| | - Yun-Long Wu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Shuaidong Huo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
4
|
Lu J, Shen X, Li H, Du J. Recent advances in bacteria-based platforms for inflammatory bowel diseases treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230142. [PMID: 39439496 PMCID: PMC11491310 DOI: 10.1002/exp.20230142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/18/2024] [Indexed: 10/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a recurring chronic inflammatory disease. Current treatment strategies are aimed at alleviating clinical symptoms and are associated with gastrointestinal or systemic adverse effects. New delivery strategies are needed for the treatment of IBD. Bacteria are promising biocarriers, which can produce drugs in situ and sense the gut in real time. Herein, we focus on recent studies of engineered bacteria used for IBD treatment and introduce the application of engineered bacteria in the diagnosis. On this basis, the current dilemmas and future developments of bacterial delivery systems are discussed.
Collapse
Affiliation(s)
- Jiaoying Lu
- Department of GastroenterologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Xinyuan Shen
- National Key Laboratory of Advanced Drug Delivery and Release SystemsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Department of BioengineeringUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release SystemsCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang UniversityHangzhouChina
| | - Juan Du
- Department of GastroenterologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
5
|
Oh SJ, Nguyen TT, Seo Y, Park HJ, Ahn JS, Shin YY, Kang BJ, Jang M, Park J, Jeong JH, Kim HS. Sustained release of stem cell secretome from nano-villi chitosan microspheres for effective treatment of atopic dermatitis. Int J Biol Macromol 2024; 277:134344. [PMID: 39089545 DOI: 10.1016/j.ijbiomac.2024.134344] [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: 11/06/2023] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Canine atopic dermatitis (AD) arises from hypersensitive immune reactions. AD symptoms entail severe pruritus and skin inflammation, with frequent relapses. Consequently, AD patients require continuous management, imposing financial burdens and mental fatigue on pet owners. In this study, we aimed to investigate the therapeutic relevance of secretome from canine adipose tissue-derived mesenchymal stem cells (MSCs), especially after encapsulation in nano-villi chitosan microspheres (CS-MS) to expect improved efficacy. Conditioned media (CM) from MSCs significantly inhibited the proliferation of splenocytes, induced the generation of regulatory T cells, and decreased mast cell degranulation. We found that beneficial soluble factors known to reduce AD symptoms, including transforming growth factor-beta 1, were detectable after sequential concentration and lyophilization of CM. The CS-MS, developed by a phase inversion regeneration method, showed high loading and sustained release of the secretome. Local injection of secretome-loaded CS-MS (ST/SC-MS) effectively reduced clinical severity compared to groups treated with secretome. Histological analysis revealed that ST/SC-MS potently suppressed epidermal hyperplasia, immunocyte infiltration and mast cell activation in the lesion. Taken together, this study presents a novel therapeutic approach exhibiting more potent and prolonged immunoregulatory efficacy of MSC secretome for canine AD treatment.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Tiep Tien Nguyen
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ye Young Shin
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Seoul 08590, Republic of Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea; BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min Jang
- Department of Veterinary Surgery, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Junhyeung Park
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
6
|
Zhao R, Yu T, Li J, Niu R, Liu D, Wang W. Single-cell encapsulation systems for probiotic delivery: Armor probiotics. Adv Colloid Interface Sci 2024; 332:103270. [PMID: 39142064 DOI: 10.1016/j.cis.2024.103270] [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: 10/31/2023] [Revised: 05/28/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Functional foods or drugs based on probiotics have gained unprecedented attention and development due to the increasingly clear relationship between probiotics and human health. Probiotics can regulate intestinal microbiota, dynamically participating in various physiological activities to directly affect human health. Some probiotic-based functional preparations have shown great potential in treating multiple refractory diseases. Currently, the survival and activity of probiotic cells in complex environments in vitro and in vivo have taken priority, and various encapsulation systems based on food-derived materials have been designed and constructed to protect and deliver probiotics. However, traditional encapsulation technology cannot achieve precise protection for a single probiotic, which makes it unable to have a significant effect after release. In this case, single-cell encapsulation systems can be assembled based on biological interfaces to protect and functionalize individual probiotic cells, maximizing their physiological activity. This review discussed the arduous challenges of probiotics in food processing, storage, human digestion, and the commonly used probiotic encapsulation system. Besides, a novel technology of probiotic encapsulation was introduced based on single-cell coating, namely, "armor probiotics". We focused on the classification, structural design, and functional characteristics of armor coatings, and emphasized the essential functional characteristics of armor probiotics in human health regulation, including regulating intestinal health and targeted bioimaging and treatment of diseased tissues. Subsequently, the benefits, limitations, potential challenges, as well as future direction of armor probiotics were put forward. We hope this review may provide new insights and ideas for developing a single-cell probiotics encapsulating system.
Collapse
Affiliation(s)
- Runan Zhao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Ting Yu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiaheng Li
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Ruihao Niu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Wenjun Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China.
| |
Collapse
|
7
|
Zhao N, Li JX, Han YJ, Lv LP, Deng J, Zhang YY. A promising strategy to improve the stability and immunogenicity of killed but metabolically active vaccines: low-temperature preparation and coating of nanoparticles. NANOSCALE 2024; 16:17118-17125. [PMID: 39189698 DOI: 10.1039/d4nr02323d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Bacteria are becoming an increasingly serious threat to human health. The emergence of super bacteria makes clinical treatment more difficult. Vaccines are one of the most effective means of preventing and treating bacterial infections. As a new class of vaccines, killed but metabolically active (KBMA) vaccines provide the immunogenicity of live vaccines and the safety of inactivated vaccines. Herein, a promising strategy is proposed to improve the stability and immunogenicity of KBMA vaccines. KBMA vaccines were produced at low temperature (4 °C), and the bacterial surface was engineered using mesoporous silica nanoparticle (MSN) coating. Compared to vaccines prepared at room temperature, the metabolic activity of KBMA vaccines prepared at 4 °C remarkably improved. Benefiting from the induction of MSNs, the stability of KBMA vaccines was increased and the preservation time was prolonged at 4 °C. Meanwhile, metabolomics analysis showed that the metabolite spectrum of live bacteria changed after photochemical treatment and MSN coating, which interfered with organic acid metabolism pathways, lipid metabolism and biosynthesis of secondary metabolites. Furthermore, the immune response in the mice treated with KBMA/MSN vaccines was similar to that in those treated with live vaccines and stronger than that in those treated with inactivated vaccines. In comparison with the control group, bacteria tissue burdens of KBMA/MSN group were significantly reduced. CD4+ T cells dominated immune responses for the protection of mice. Thus, the current work promotes the application of KBMA vaccines, providing an alternative choice for treating bacterial infections.
Collapse
Affiliation(s)
- Ning Zhao
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Jia-Xv Li
- Academy of Military Medical Sciences, Beijing 100850, China.
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yong-Jiao Han
- Academy of Military Medical Sciences, Beijing 100850, China.
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Li-Ping Lv
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Jiang Deng
- Academy of Military Medical Sciences, Beijing 100850, China.
| | - Yan-Yu Zhang
- Academy of Military Medical Sciences, Beijing 100850, China.
| |
Collapse
|
8
|
Zheng B, Li M, Zhang T, Li B, Li Q, Saiding Q, Chen W, Guo M, Koo S, Ji X, Tao W. Functional modification of gut bacteria for disease diagnosis and treatment. MED 2024; 5:863-885. [PMID: 38964334 DOI: 10.1016/j.medj.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/15/2023] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Intestinal bacteria help keep humans healthy by regulating lipid and glucose metabolism as well as the immunological and neurological systems. Oral treatment using intestinal bacteria is limited by the high acidity of stomach fluids and the immune system's attack on foreign bacteria. Scientists have created coatings and workarounds to overcome these limitations and improve bacterial therapy. These preparations have demonstrated promising outcomes, with advances in synthetic biology and optogenetics improving their focused colonization and controlled release. Engineering bacteria preparations have become a revolutionary therapeutic approach that converts intestinal bacteria into cellular factories for medicinal chemical synthesis. The present paper discusses various aspects of engineering bacteria preparations, including wrapping materials, biomedical uses, and future developments.
Collapse
Affiliation(s)
- Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Mengyi Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Tiange Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Bowen Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qiuya Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mingming Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Li C, Wang ZX, Xiao H, Wu FG. Intestinal Delivery of Probiotics: Materials, Strategies, and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310174. [PMID: 38245861 DOI: 10.1002/adma.202310174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/04/2024] [Indexed: 01/22/2024]
Abstract
Probiotics with diverse and crucial properties and functions have attracted broad interest from many researchers, who adopt intestinal delivery of probiotics to modulate the gut microbiota. However, the major problems faced for the therapeutic applications of probiotics are the viability and colonization of probiotics during their processing, oral intake, and subsequent delivery to the gut. The challenges of simple oral delivery (stability, controllability, targeting, etc.) have greatly limited the use of probiotics in clinical therapies. Nanotechnology can endow the probiotics to be delivered to the intestine with improved survival rate and increased resistance to the adverse environment. Additionally, the progress in synthetic biology has created new opportunities for efficiently and purposefully designing and manipulating the probiotics. In this article, a brief overview of the types of probiotics for intestinal delivery, the current progress of different probiotic encapsulation strategies, including the chemical, physical, and genetic strategies and their combinations, and the emerging single-cell encapsulation strategies using nanocoating methods, is presented. The action mechanisms of probiotics that are responsible for eliciting beneficial effects are also briefly discussed. Finally, the therapeutic applications of engineered probiotics are discussed, and the future trends toward developing engineered probiotics with advanced features and improved health benefits are proposed.
Collapse
Affiliation(s)
- Chengcheng Li
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Zi-Xi Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
10
|
Gao P, Duan Z, Xu G, Gong Q, Wang J, Luo K, Chen J. Harnessing and Mimicking Bacterial Features to Combat Cancer: From Living Entities to Artificial Mimicking Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405075. [PMID: 39136067 DOI: 10.1002/adma.202405075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Bacterial-derived micro-/nanomedicine has garnered considerable attention in anticancer therapy, owing to the unique natural features of bacteria, including specific targeting ability, immunogenic benefits, physicochemical modifiability, and biotechnological editability. Besides, bacterial components have also been explored as promising drug delivery vehicles. Harnessing these bacterial features, cutting-edge physicochemical and biotechnologies have been applied to attenuated tumor-targeting bacteria with unique properties or functions for potent and effective cancer treatment, including strategies of gene-editing and genetic circuits. Further, the advent of bacteria-inspired micro-/nanorobots and mimicking artificial systems has furnished fresh perspectives for formulating strategies for developing highly efficient drug delivery systems. Focusing on the unique natural features and advantages of bacteria, this review delves into advances in bacteria-derived drug delivery systems for anticancer treatment in recent years, which has experienced a process from living entities to artificial mimicking systems. Meanwhile, a summary of relative clinical trials is provided and primary challenges impeding their clinical application are discussed. Furthermore, future directions are suggested for bacteria-derived systems to combat cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Gang Xu
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kui Luo
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jie Chen
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
11
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Fan Y, Ye J, Kang Y, Niu G, Shi J, Yuan X, Li R, Han J, Ji X. Biomimetic piezoelectric nanomaterial-modified oral microrobots for targeted catalytic and immunotherapy of colorectal cancer. SCIENCE ADVANCES 2024; 10:eadm9561. [PMID: 38718119 PMCID: PMC11078194 DOI: 10.1126/sciadv.adm9561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Lactic acid (LA) accumulation in the tumor microenvironment poses notable challenges to effective tumor immunotherapy. Here, an intelligent tumor treatment microrobot based on the unique physiological structure and metabolic characteristics of Veillonella atypica (VA) is proposed by loading Staphylococcus aureus cell membrane-coating BaTiO3 nanocubes (SAM@BTO) on the surface of VA cells (VA-SAM@BTO) via click chemical reaction. Following oral administration, VA-SAM@BTO accurately targeted orthotopic colorectal cancer through inflammatory targeting of SAM and hypoxic targeting of VA. Under in vitro ultrasonic stimulation, BTO catalyzed two reduction reactions (O2 → •O2- and CO2 → CO) and three oxidation reactions (H2O → •OH, GSH → GSSG, and LA → PA) simultaneously, effectively inducing immunogenic death of tumor cells. BTO catalyzed the oxidative coupling of VA cells metabolized LA, effectively disrupting the immunosuppressive microenvironment, improving dendritic cell maturation and macrophage M1 polarization, and increasing effector T cell proportions while decreasing regulatory T cell numbers, which facilitates synergetic catalysis and immunotherapy.
Collapse
Affiliation(s)
- Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Jiamin Ye
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Yong Kang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Gaoli Niu
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Jiacheng Shi
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Xue Yuan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Jingwen Han
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Medical College, Linyi University, Linyi 276000, China
| |
Collapse
|
13
|
Han J, McClements DJ, Liu X, Liu F. Oral delivery of probiotics using single-cell encapsulation. Compr Rev Food Sci Food Saf 2024; 23:e13322. [PMID: 38597567 DOI: 10.1111/1541-4337.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Adequate intake of live probiotics is beneficial to human health and wellbeing because they can help treat or prevent a variety of health conditions. However, the viability of probiotics is reduced by the harsh environments they experience during passage through the human gastrointestinal tract (GIT). Consequently, the oral delivery of viable probiotics is a significant challenge. Probiotic encapsulation provides a potential solution to this problem. However, the production methods used to create conventional encapsulation technologies often damage probiotics. Moreover, the delivery systems produced often do not have the required physicochemical attributes or robustness for food applications. Single-cell encapsulation is based on forming a protective coating around a single probiotic cell. These coatings may be biofilms or biopolymer layers designed to protect the probiotic from the harsh gastrointestinal environment, enhance their colonization, and introduce additional beneficial functions. This article reviews the factors affecting the oral delivery of probiotics, analyses the shortcomings of existing encapsulation technologies, and highlights the potential advantages of single-cell encapsulation. It also reviews the various approaches available for single-cell encapsulation of probiotics, including their implementation and the characteristics of the delivery systems they produce. In addition, the mechanisms by which single-cell encapsulation can improve the oral bioavailability and health benefits of probiotics are described. Moreover, the benefits, limitations, and safety issues of probiotic single-cell encapsulation technology for applications in food and beverages are analyzed. Finally, future directions and potential challenges to the widespread adoption of single-cell encapsulation of probiotics are highlighted.
Collapse
Affiliation(s)
- Jiaqi Han
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| | - David Julian McClements
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| |
Collapse
|
14
|
Santos‐Beneit F. What is the role of microbial biotechnology and genetic engineering in medicine? Microbiologyopen 2024; 13:e1406. [PMID: 38556942 PMCID: PMC10982607 DOI: 10.1002/mbo3.1406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
Microbial products are essential for developing various therapeutic agents, including antibiotics, anticancer drugs, vaccines, and therapeutic enzymes. Genetic engineering techniques, functional genomics, and synthetic biology unlock previously uncharacterized natural products. This review highlights major advances in microbial biotechnology, focusing on gene-based technologies for medical applications.
Collapse
Affiliation(s)
- Fernando Santos‐Beneit
- Institute of Sustainable ProcessesValladolidSpain
- Department of Chemical Engineering and Environmental Technology, School of Industrial EngineeringUniversity of ValladolidValladolidSpain
| |
Collapse
|
15
|
Niu L, Liu Y, Li N, Wang Y, Kang L, Su X, Xu C, Sun Z, Sang W, Xu J, Guo H, Shen S. Oral probiotics microgel plus Galunisertib reduced TGF-β blockade resistance and enhanced anti-tumor immune responses in colorectal cancer. Int J Pharm 2024; 652:123810. [PMID: 38244648 DOI: 10.1016/j.ijpharm.2024.123810] [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: 10/05/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Transforming growth factor β (TGF-β), a versatile immunosuppressive cytokine, has gained increasing attention as a potential target for cancer immunotherapy. However, current strategies are constrained by tumor heterogeneity and drug resistance. Therapeutic probiotics, such as Escherichia coli Nissle1917 (EcN), not only regulate the gut microbiota to increase beneficial bacteria with anti-tumor effects, but also modulate immune factors within the body, thereby enhancing immunity. In this study, we developed an oral microgel delivery system of EcN@(CS-SA)2 by electrostatic interaction between chitosan (CS) and sodium alginate (SA), aiming to enhance its bioavailability in the gastrointestinal tract (GIT). Notably, EcN@(CS-SA)2 microgel showed a synergistic enhancement of the anti-tumor efficacy of Galunisertib (Gal, a TGF-β inhibitor) by inducing apoptosis and immunogenic cell death (ICD) in tumor cells, as well as promoting increased infiltration of CD8+ T cells into the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Lili Niu
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yao Liu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Nannan Li
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yang Wang
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Lin Kang
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiaomin Su
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Ce Xu
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Zanya Sun
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China; Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Weicong Sang
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Jingyuan Xu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China.
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| |
Collapse
|
16
|
Yan C, Kim SR. Microencapsulation for Pharmaceutical Applications: A Review. ACS APPLIED BIO MATERIALS 2024; 7:692-710. [PMID: 38320297 DOI: 10.1021/acsabm.3c00776] [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: 02/08/2024]
Abstract
In order to improve bioavailability, stability, control release, and target delivery of active pharmaceutical ingredients (APIs), as well as to mask their bitter taste, to increase their efficacy, and to minimize their side effects, a variety of microencapsulation (including nanoencapsulation, particle size <100 nm) technologies have been widely used in the pharmaceutical industry. Commonly used microencapsulation technologies are emulsion, coacervation, extrusion, spray drying, freeze-drying, molecular inclusion, microbubbles and microsponge, fluidized bed coating, supercritical fluid encapsulation, electro spinning/spray, and polymerization. In this review, APIs are categorized by their molecular complexity: small APIs (compounds with low molecular weight, like Aspirin, Ibuprofen, and Cannabidiol), medium APIs (compounds with medium molecular weight like insulin, peptides, and nucleic acids), and living microorganisms (such as probiotics, bacteria, and bacteriophages). This article provides an overview of these microencapsulation technologies including their processes, matrix, and their recent applications in microencapsulation of APIs. Furthermore, the advantages and disadvantages of these common microencapsulation technologies in terms of improving the efficacy of APIs for pharmaceutical treatments are comprehensively analyzed. The objective is to summarize the most recent progresses on microencapsulation of APIs for enhancing their bioavailability, control release, target delivery, masking their bitter taste and stability, and thus increasing their efficacy and minimizing their side effects. At the end, future perspectives on microencapsulation for pharmaceutical applications are highlighted.
Collapse
Affiliation(s)
- Cuie Yan
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| | - Sang-Ryoung Kim
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| |
Collapse
|
17
|
Cao Z, Pang Y, Pu J, Liu J. Bacteria-based drug delivery for treating non-oncological diseases. J Control Release 2024; 366:668-683. [PMID: 38219912 DOI: 10.1016/j.jconrel.2024.01.020] [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: 11/19/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
Bacteria inhabit all over the human body, especially the skin, gastrointestinal tract, respiratory tract, urogenital tract, as well as specific lesion sites, such as wound and tumor. By leveraging their distinctive attributes including rapid proliferation, inherent abilities to colonize various biointerfaces in vivo and produce diverse biomolecules, and the flexibility to be functionalized via genetic engineering or surface modification, bacteria have been widely developed as living therapeutic agents, showing promising potential to make a great impact on the exploration of advanced drug delivery systems. In this review, we present an overview of bacteria-based drug delivery and its applications in treating non-oncological diseases. We systematically summarize the physiological positions where living bacterial therapeutic agents can be delivered to, including the skin, gastrointestinal tract, respiratory tract, and female genital tract. We discuss the success of using bacteria-based drug delivery systems in the treatment of diseases that occur in specific locations, such as skin wound healing/infection, inflammatory bowel disease, respiratory diseases, and vaginitis. We also discuss the advantages as well as the limitations of these living therapeutics and bacteria-based drug delivery, highlighting the key points that need to be considered for further translation. This review article may provide unique insights for designing next-generation bacteria-based therapeutics and developing advanced drug delivery systems.
Collapse
Affiliation(s)
- Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yan Pang
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
18
|
Guo C, Liu J, Zhang Y. Current advances in bacteria-based cancer immunotherapy. Eur J Immunol 2024; 54:e2350778. [PMID: 38105295 DOI: 10.1002/eji.202350778] [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: 09/17/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
As the understanding of the tumor microenvironment has deepened, immunotherapy has become a promising strategy for cancer treatment. In contrast to traditional therapies, immunotherapy is more precise and induces fewer adverse effects. In this field, some bacteria have attracted increased attention because of their natural ability to preferentially colonize and proliferate inside tumor sites and exert antitumor effects. Moreover, bacterial components may activate innate and adaptive immunity to resist tumor progression. However, the application of bacteria-based cancer immunotherapy is hampered by potential infection-associated toxicity and unpredictable behavior in vivo. Owing to modern developments in genetic engineering, bacteria can be modified to weaken their toxicity and enhance their ability to eliminate tumor cells or activate the antitumor immune response. This review summarizes the roles of bacteria in the tumor microenvironment, current strategies for bacterial engineering, and the synergistic efficiency of bacteria with other immunotherapies. In addition, the prospects and challenges of the clinical translation of engineered bacteria are summarized.
Collapse
Affiliation(s)
- Caijuan Guo
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinyan Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & and Treatment, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Han K, Xu J, Xie F, Crowther J, Moon JJ. Engineering Strategies to Modulate the Gut Microbiome and Immune System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:208-215. [PMID: 38166246 PMCID: PMC10766079 DOI: 10.4049/jimmunol.2300480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/28/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiota, predominantly residing in the colon, is a complex ecosystem with a pivotal role in the host immune system. Dysbiosis of the gut microbiota has been associated with various diseases, and there is an urgent need to develop new therapeutics that target the microbiome and restore immune functions. This Brief Review discusses emerging therapeutic strategies that focus on oral delivery systems for modulating the gut microbiome. These strategies include genetic engineering of probiotics, probiotic-biomaterial hybrids, dietary fibers, and oral delivery systems for microbial metabolites, antimicrobial peptides, RNA, and antibiotics. Engineered oral formulations have demonstrated promising outcomes in reshaping the gut microbiome and influencing immune responses in preclinical studies. By leveraging these approaches, the interplay between the gut microbiota and the immune system can be harnessed for the development of novel therapeutics against cancer, autoimmune disorders, and allergies.
Collapse
Affiliation(s)
- Kai Han
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fang Xie
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Julia Crowther
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Abdi F, Buzhor MG, Zellweger N, Zhi-Luo, Leroux JC. pH-dependent pressure-sensitive colonic capsules for the delivery of aqueous bacterial suspensions. J Control Release 2024; 365:688-702. [PMID: 38040343 DOI: 10.1016/j.jconrel.2023.11.048] [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: 06/14/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
Microbiome-based therapies hold great promise for treating various diseases, but the efficient delivery of live bacteria to the colon remains a challenge. Furthermore, current oral formulations, such as lyophilized bacterial capsules or tablets, are produced using processes that can decrease bacterial viability. Consequently, high dosages are required to achieve efficacy. Herein, we report the design of pressure-sensitive colonic capsules for the encapsulation and delivery of aqueous suspensions of live bacteria. The capsules consisted of 2 functional thin-films (hydrophobic and enteric) of ethyl cellulose and Eudragit S100 dip-coated onto hydroxypropyl methylcellulose molds. The capsules could be loaded with aqueous media and provide protection against acidic fluids and, to some extent, oxygen diffusion, suggesting their potential suitability for delivering anaerobic bacterial strains. Disintegration and mechanical studies indicated that the capsules could withstand transit through the stomach and upper/proximal small intestinal segments and rupture in the ileum/colon. In vitro studies showed that bacterial cells (anaerobic and aerobic commensals) remained highly viable (74-98%) after encapsulation and exposure to the simulated GI tract conditions. In vivo studies with a beagle dog model revealed that 67% of the capsules opened after 3.5 h, indicating content release in the distal gastrointestinal tract. These data demonstrate that live aqueous bacterial suspensions comprised of both aerobic and anaerobic commensals can be encapsulated and in the future might be efficiently delivered to the distal gastrointestinal tract, suggesting the practical applications of these capsules in microbiome-based therapies.
Collapse
Affiliation(s)
- Fatma Abdi
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Marina Green Buzhor
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Nadia Zellweger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Zhi-Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, 518055 Shenzhen, Guangdong, China
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
21
|
Lin Q, Si Y, Zhou F, Hao W, Zhang P, Jiang P, Cha R. Advances in polysaccharides for probiotic delivery: Properties, methods, and applications. Carbohydr Polym 2024; 323:121414. [PMID: 37940247 DOI: 10.1016/j.carbpol.2023.121414] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 11/10/2023]
Abstract
Probiotics are essential to improve the health of the host, whereas maintaining the viability of probiotics in harsh environments remains a challenge. Polysaccharides have non-toxicity, excellent biocompatibility, and outstanding biodegradability, which can protect probiotics by forming a physical barrier and show a promising prospect for probiotic delivery. In this review, we summarize polysaccharides commonly used for probiotic microencapsulation and introduce the microencapsulation technologies, including extrusion, emulsion, spray drying, freeze drying, and electrohydrodynamics. We discuss strategies for better protection of probiotics and introduce the applications of polysaccharides-encapsulated probiotics in functional food, oral formulation, and animal feed. Finally, we propose the challenges of polysaccharides-based delivery systems in industrial production and application. This review will help provide insight into the advances and challenges of polysaccharides in probiotic delivery.
Collapse
Affiliation(s)
- Qianqian Lin
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing 100083, PR China; Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, PR China.
| | - Yanxue Si
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing 100083, PR China.
| | - Fengshan Zhou
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing 100083, PR China.
| | - Wenshuai Hao
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing 100083, PR China.
| | - Pai Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology, China University of Geosciences (Beijing), No. 29 Xueyuan Road, Haidian District, Beijing 100083, PR China.
| | - Peng Jiang
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, PR China; College of Chemistry and Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Ruitao Cha
- Laboratory of Theoretical and Computational Nanoscience, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11 Zhongguancun Beiyitiao, Haidian District, Beijing 100190, PR China.
| |
Collapse
|
22
|
Zheng BD, Gan L, Tian LY, Chen GH. Protein/polysaccharide-based hydrogels loaded probiotic-mediated therapeutic systems: A review. Int J Biol Macromol 2023; 253:126841. [PMID: 37696368 DOI: 10.1016/j.ijbiomac.2023.126841] [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: 08/01/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
The natural characteristics of protein/polysaccharide-based hydrogels, as a potential drug delivery platform, have attracted extensive attention. Probiotics have attracted renewed interest in drug research because of their beneficial effects on host health. The idea of using probiotics loaded on protein/polysaccharide-based hydrogels as potential drugs to treat different diseases has been put forward and shows great prospects. Based on this, in this review, we highlight the design strategy of hydrogels loaded probiotic-mediated therapy systems and review the potential diseases that have been proved to be treatable in the laboratory, including promoting wound healing and improving intestinal health and vaginal health, and discuss the challenges existing in the current design.
Collapse
Affiliation(s)
- Bing-De Zheng
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Lei Gan
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Li-Yuan Tian
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Guan-Hong Chen
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
23
|
Lin S, Wu F, Zhang Y, Chen H, Guo H, Chen Y, Liu J. Surface-modified bacteria: synthesis, functionalization and biomedical applications. Chem Soc Rev 2023; 52:6617-6643. [PMID: 37724854 DOI: 10.1039/d3cs00369h] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
The past decade has witnessed a great leap forward in bacteria-based living agents, including imageable probes, diagnostic reagents, and therapeutics, by virtue of their unique characteristics, such as genetic manipulation, rapid proliferation, colonization capability, and disease site targeting specificity. However, successful translation of bacterial bioagents to clinical applications remains challenging, due largely to their inherent susceptibility to environmental insults, unavoidable toxic side effects, and limited accumulation at the sites of interest. Cell surface components, which play critical roles in shaping bacterial behaviors, provide an opportunity to chemically modify bacteria and introduce different exogenous functions that are naturally unachievable. With the help of surface modification, a wide range of functionalized bacteria have been prepared over the past years and exhibit great potential in various biomedical applications. In this article, we mainly review the synthesis, functionalization, and biomedical applications of surface-modified bacteria. We first introduce the approaches of chemical modification based on the bacterial surface structure and then highlight several advanced functions achieved by modifying specific components on the surface. We also summarize the advantages as well as limitations of surface chemically modified bacteria in the applications of bioimaging, diagnosis, and therapy and further discuss the current challenges and possible solutions in the future. This work will inspire innovative design thinking for the development of chemical strategies for preparing next-generation biomedical bacterial agents.
Collapse
Affiliation(s)
- Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yifan Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Huan Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
24
|
Golchin A, Ranjbarvan P, Parviz S, Shokati A, Naderi R, Rasmi Y, Kiani S, Moradi F, Heidari F, Saltanatpour Z, Alizadeh A. The role of probiotics in tissue engineering and regenerative medicine. Regen Med 2023; 18:635-657. [PMID: 37492007 DOI: 10.2217/rme-2022-0209] [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: 07/27/2023] Open
Abstract
Tissue engineering and regenerative medicine (TERM) as an emerging field is a multidisciplinary science and combines basic sciences such as biomaterials science, biology, genetics and medical sciences to achieve functional TERM-based products to regenerate or replace damaged or diseased tissues or organs. Probiotics are useful microorganisms which have multiple effective functions on human health. They have some immunomodulatory and biocompatibility effects and improve wound healing. In this article, we describe the latest findings on probiotics and their pro-healing properties on various body systems that are useable in regenerative medicine. Therefore, this review presents a new perspective on the therapeutic potential of probiotics for TERM.
Collapse
Affiliation(s)
- Ali Golchin
- Cellular & Molecular Research Center, Cellular & Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
- Department of Clinical Biochemistry & Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
| | - Parviz Ranjbarvan
- Cellular & Molecular Research Center, Cellular & Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
- Department of Clinical Biochemistry & Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
| | - Shima Parviz
- Department of Tissue Engineering & Applied cell sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, 71348-14336, Iran
| | - Amene Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Roya Naderi
- Neurophysiology Research center & Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
| | - Yousef Rasmi
- Cellular & Molecular Research Center & Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, 57157993313, Iran
| | - Samaneh Kiani
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, 48157-33971, Iran
| | - Faezeh Moradi
- Department of Tissue engineering, Medical Sciences Faculty, Tarbiat Modares University, Tehran, 14117-13116, Iran
| | - Fahimeh Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, 71348-14336, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, 71348-14336, Iran
| | - Zohreh Saltanatpour
- Pediatric Cell & Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Stem Cell & Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Akram Alizadeh
- Nervous System Stem Cells Research Center & Department of Tissue Engineering & Applied Cell Sciences, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, 35147-99422, Iran
| |
Collapse
|
25
|
Chen J, Zhang P, Wu C, Yao Q, Cha R, Gao Y. Reductase-Labile Peptidic Supramolecular Hydrogels Aided in Oral Delivery of Probiotics. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37339324 DOI: 10.1021/acsami.3c04408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Oral delivery of probiotics has been a promising method for treatment of inflammatory bowel diseases (IBDs). However, probiotics always suffer from substantial loss of viability due to the harsh gastrointestinal conditions, especially the highly acidic environment in the stomach and bile salts in the intestine. In addition, to overcome the challenging conditions, an ideal delivery of probiotics requires the on-demand release of probiotics upon environmental response. Herein, a novel nitroreductase (NTR) labile peptidic hydrogel based on supramolecular self-assembly is demonstrated. The efficient encapsulation of typical probiotic Escherichia coli Nissle 1917 (EcN) into supramolecular assemblies yielded a probiotic-loaded hydrogel (EcN@Gel). Such a hydrogel adequately protected EcN to improve its viability against harsh acid and bile salt environments during oral delivery. The upregulated NTR in the intestinal tract triggered the disassembly of the hydrogel and accomplished the controlled release of EcN locally. In ulcerative colitis (UC)-bearing mice, EcN@Gel showed significantly enhanced therapeutic efficacy by downregulating proinflammatory cytokines and repairing the intestinal barrier. Moreover, EcN@Gel remolded the gut microbiome by increasing the diversity and abundance of indigenous probiotics, contributing to ameliorated therapies of IBDs. The NTR-labile hydrogel provided a promising platform for the on-demand delivery of probiotics into the intestinal tract.
Collapse
Affiliation(s)
- Jiali Chen
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pai Zhang
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Chengling Wu
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Qingxin Yao
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Ruitao Cha
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuan Gao
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
26
|
Jiang X, Yan C, Zhang H, Chen L, Jiang R, Zheng K, Jin W, Ma H, Liu X, Dong M. Oral Probiotic Expressing Human Ethanol Dehydrogenase Attenuates Damage Caused by Acute Alcohol Consumption in Mice. Microbiol Spectr 2023; 11:e0429422. [PMID: 37039510 PMCID: PMC10269551 DOI: 10.1128/spectrum.04294-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/24/2023] [Indexed: 04/12/2023] Open
Abstract
Alcohol is an essential drug in human life with multiple medical functions, but excessive alcohol intake, even a single episode of binge drinking, can cause serious damage. Reducing alcohol consumption or absorption is a direct way to alleviate the related harm. Alcohol is decomposed successively by alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH) in the liver. Here, we produced a human ADH1B (hADH1B)-expressing probiotic, a recombinant Lactococcus lactis, that aimed to enhance alcohol degradation in the intestinal tract after oral administration. Our results showed that the oral hADH1B-expressing probiotic reduced alcohol absorption, prolonged the alcohol tolerance time, and shortened the recovery time after acute alcohol challenge. More importantly, the liver and intestine were protected from acute injury caused by alcohol challenge. Therefore, the engineered probiotic has the potential to protect organ damage from alcohol consumption. Furthermore, this engineered probiotic may have beneficial effects on alcohol-related diseases such as alcoholic fatty liver disease. IMPORTANCE Alcohol plays an important role in medical treatment, culture, and social interaction. However, excessive alcohol consumption or improper alcohol intake patterns can lead to serious damage to health. Aiming to reduce the harm of alcohol consumption, we designed a recombinant probiotic expressing hADH1B. Our results showed that this recombinant probiotic can reduce alcohol absorption and protect the body from alcohol damage, including hangover, liver, and intestinal damage. Reducing alcohol damage is helpful to the health of people with difficulty in abstinence. The engineered probiotic may provide new strategies for treatment and prevention of the negative effects of alcohol, and it also has the potential for widespread application.
Collapse
Affiliation(s)
- Xiaoxiao Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chunlong Yan
- Agriculture College of Yanbian University, Yanji, Jilin, China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Li Chen
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Rui Jiang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Kexin Zheng
- Institute of Infectious Disease, Ditan Hospital, Capital Medical University, Beijing, China
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Huijuan Ma
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaomeng Liu
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Hirano-Kusuda M, Setoguchi S, Koga M, Goto S, Yamada A, Watase D, Nagata-Akaho N, Karube Y, Matsunaga K, Takata J. Cationic Ester Prodrugs of Curcumin with N,N-dimethyl Amino Acid Promoieties Improved Poor Water Solubility and Intestinal Absorption. Pharm Res 2023; 40:1299-1310. [PMID: 37081301 DOI: 10.1007/s11095-023-03500-5] [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: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE Although curcumin (Cur) has powerful pharmacological effects, its use in medicine has not been established yet. The oral bioavailability (BA) of Cur is limited because of its poor water solubility. The purpose of this study was to confirm whether cationic N,N-dimethyl amino acid esters of Cur could act as prodrugs and improve its water solubility and oral bioavailability. METHODS Two N,N-dimethyl amino acid esters of Cur were synthesized. The hydrolysis profile of the esters was evaluated using rat and human microsomes. A pharmacokinetic study after oral administration of the Cur ester derivatives was performed in rats and compared to the administration of suspended or dissolved Cur formulation. The anti-inflammatory effects of the Cur derivatives were evaluated using macrophage RAW 264.7 stimulated with lipopolysaccharide. RESULTS Cur ester derivatives showed > 200 mM water solubility. The derivatives were reconverted to the parent compound (Cur) after cleavage of the ester bonds by microsomal esterase, indicating that the compounds could act as Cur prodrugs. The Cur prodrugs enhanced the absolute oral bioavailability of Cur by a 9- and threefold increase of suspended and dissolved Cur administration, respectively, thereby improving intestinal absorption. Cur prodrugs strongly attenuated COX2, iNOS, and ERK phosphorylation. CONCLUSIONS The cationic N,N-dimethyl amino acid ester prodrugs of Cur improved the water solubility of Cur and enhanced oral bioavailability in rats. These Cur prodrugs may be good candidates for developing medicinal options previously unavailable due to the poor water solubility and oral BA of Cur.
Collapse
Affiliation(s)
- Mariko Hirano-Kusuda
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shuichi Setoguchi
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Mitsuhisa Koga
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shotaro Goto
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Ayano Yamada
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Daisuke Watase
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Nami Nagata-Akaho
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yoshiharu Karube
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kazuhisa Matsunaga
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Jiro Takata
- Faculty of Pharmaceutical Sciences, Fukuoka University, Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
28
|
Wang J, Ghosh D, Maniruzzaman M. Using bugs as drugs: administration of bacteria-related microbes to fight cancer. Adv Drug Deliv Rev 2023; 197:114825. [PMID: 37075953 DOI: 10.1016/j.addr.2023.114825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Driven by the advancement of microbiology and cancer biology, bioengineering of bacteria-related microbes has demonstrated great potential in targeted cancer therapy. Presently, the major administration routes of bacteria-related microbes for cancer treatment include intravenous injection, intratumoral injection, intraperitoneal injection, and oral delivery. Administration routes of bacteria play a key role in anticancer therapeutic efficacy since different delivery approaches might exert an anticancer effect through diverse mechanisms. Herein, we provide an overview of the primary routes of bacteria administration as well as their advantages and limitations. Furthermore, we discuss that microencapsulation can overcome the current challenges of direct administration of free bacteria. We also review the latest advancements in combining functional particles with engineered bacteria to fight against cancer, which can be further coupled with conventional anticancer therapies to improve the therapeutic effect. Eventually, we highlight the application prospect of bioprinting in cancer bacteriotherapy, which enables the long-term sustained delivery and individualized dose regimen, representing a new paradigm for personalized cancer treatment.
Collapse
Affiliation(s)
- Jiawei Wang
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
29
|
Xie X, Li Q, Jia L, Yuan H, Guo T, Meng T. Multishell Colloidosome Platform with Sequential Gastrointestinal Resistance for On-Demand Probiotic Delivery. Adv Healthc Mater 2023; 12:e2202954. [PMID: 36652659 DOI: 10.1002/adhm.202202954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/28/2022] [Indexed: 01/19/2023]
Abstract
Probiotic-based oral therapy can potentially prevent and treat diseases by regulating the balance of intestinal flora. However, significant loss of viability and bioactivity of probiotics before reaching the colon results in low delivery efficiency and therapeutic effects, which limits their clinical applications. Here, this work proposes a multishell colloidosome (MSC) platform with sequential gastrointestinal resistance for on-demand probiotic delivery based on biomimetic mineralization and microfluidic technology. Notably, the viability of the decorated probiotics increases 280-fold compared to that of free bacteria during preservation. Because of the sequential gastrointestinal resistance of MSC, encapsulated probiotics exhibit high viability (61%) under continuous exposure to extreme acidity, bile salt erosion, and enzymatic action, whereas free bacteria have a viability of 0%. Moreover, in vitro and in vivo studies reveal that MSC mainly releases probiotics in the colon and improves colonic colonization by probiotics to maintain the integrity of the intestinal barrier and regulate the balance of intestinal flora. Consequently, MSC significantly improves the therapeutic effect on colitis in mice. The MSC platform provides a promising delivery strategy to enhance the efficacy of orally administered probiotics.
Collapse
Affiliation(s)
- Xin Xie
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Qinyuan Li
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Lufan Jia
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Hao Yuan
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Ting Guo
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Tao Meng
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| |
Collapse
|
30
|
Jeong Y, Kong W, Lu T, Irudayaraj J. Soft hydrogel-shell confinement systems as bacteria-based bioactuators and biosensors. Biosens Bioelectron 2023; 219:114809. [PMID: 36274428 DOI: 10.1016/j.bios.2022.114809] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/25/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Genetically engineered (GE) bacteria were utilized for developing functional systems upon confinement within a restricted space. Use of natural soft hydrogel such as alginate, gelatin, and agarose, have been investigated as promising approaches to design functional architectures. Nevertheless, a challenge is to develop functional microenvironments that support biofilm-like confinement in a relevant three-dimensional (3D) format for long-term studies. We demonstrate a natural soft hydrogel bioactuator based on alginate core-shell structures (0.25-2 mm core and 50-300 μm shell thickness) that enables 3D microbial colonization upon confinement with high cell density. Specially, our study evaluates the efficiency of bacteria-functional system by recapitulating various GE bacteria which can produce common reporter proteins, to demonstrate their actuator functions as well as dynamic pair-wise interactions. The structural design of the hydrogel can endure continued growth of various bacteria colonies within the confined space for over 10 days. The total amount of cellular biomass upon hydrogel-shell confinement was increased 5-fold compared to conventional techniques without hydrogel-shell. Furthermore, the enzymatic activity increased 3.8-fold and bioluminescence signal by 8-fold compared to the responses from conventional hydrogel systems. The conceptualized platform and our workflow represent a reliable strategy with core-shell structures to develop artificial hydrogel habitats as bacteria-based functional systems for bioactuation.
Collapse
Affiliation(s)
- Yoon Jeong
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Wentao Kong
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology and Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ting Lu
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology and Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joseph Irudayaraj
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology and Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
31
|
Zhang L, Zhao M, Fu X. Gastric microbiota dysbiosis and Helicobacter pylori infection. Front Microbiol 2023; 14:1153269. [PMID: 37065152 PMCID: PMC10098173 DOI: 10.3389/fmicb.2023.1153269] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/14/2023] [Indexed: 04/18/2023] Open
Abstract
Helicobacter pylori (H. pylori) infection is one of the most common causes of gastric disease. The persistent increase in antibiotic resistance worldwide has made H. pylori eradication challenging for clinicians. The stomach is unsterile and characterized by a unique niche. Communication among microorganisms in the stomach results in diverse microbial fitness, population dynamics, and functional capacities, which may be positive, negative, or neutral. Here, we review gastric microecology, its imbalance, and gastric diseases. Moreover, we summarize the relationship between H. pylori and gastric microecology, including non-H. pylori bacteria, fungi, and viruses and the possibility of facilitating H. pylori eradication by gastric microecology modulation, including probiotics, prebiotics, postbiotics, synbiotics, and microbiota transplantation.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Gastroenterology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Zhang X, Zhang Y, Wang N, Shen Y, Chen Q, Han L, Hu B. Photothermal Nanoheaters-Modified Spores for Safe and Controllable Antitumor Therapy. Int J Nanomedicine 2022; 17:6399-6412. [PMID: 36545219 PMCID: PMC9762999 DOI: 10.2147/ijn.s385269] [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: 08/11/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction To present a safer tumor therapy based on bacteria and identify in detail how the activation and infection behavior of spores can be controlled remotely by near-infrared light (NIR-irradiation) based on nanoheaters' modification. Methods Spores bring a better tolerance to surface modification. Transitive gold-nanorods-allied-nanoclusters-modified spores (Spore@NRs/NCs) were constructed by covalent glutaraldehyde crosslink. The photothermal properties of nanoheaters before and after attachment to spores were studied by recording temperature-irradiation time curves. The controlled viability and infection behavior of Spore@NRs/NCs were investigated by NIR-irradiation. Results In this work, a controllable sterilizing effect to activated vegetative bacteria was obtained obviously. When met with a suitable growth-environment, Spore@NRs/NCs could germinate, activate into vegetative bacteria and continue to reproduce. Without NIR-irradiation, nanoheaters could not affect the activity of both spores and vegetative bacterial cells. However, with NIR-irradiation after incubating in growth medium, nanoheaters on spores could control the spores' germination and affect the growth curve as well as the viability of the vegetative bacterial cells. For Spore@NRs/NCs (Spore:NCs:NRs=1:1:4, 67.5 μg mL-1), a ~98% killing rate of vegetative bacterial cells was obtained with NIR-irradiation (2.8 W cm-2, 20 min) after 2 h-incubation. In addition, these nanoheaters modified on spores could be taken not only to the vegetative bacteria cells, but also to the first-generation bacteria cells with their excellent photothermal and bactericidal performance, as well as synergetic anticancer effect. NIR-irradiation after 2 h-incubation could also trigger Spore@NRs/NCs (1:1:4, 6 μL) to synergistically reduce the viability of HCT116 cells to 15.63±2.90%. Conclusion By using NIR-irradiation, the "transitive" nanoheaters can remotely control the activity of both bacteria (germinated from spore) and cancer cells. This discovery provides basis and a feasible plan for controllable safer treatment of bacteria therapy, especially anaerobes with spores in hypoxic areas of the malignant solid tumors.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, People’s Republic of China
| | - Yang Zhang
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, People’s Republic of China
| | - Ning Wang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, People’s Republic of China
| | - Yetong Shen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, People’s Republic of China
| | - Qing Chen
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, People’s Republic of China
| | - Lu Han
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, People’s Republic of China
| | - Bo Hu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, People’s Republic of China,Correspondence: Bo Hu, Email ;
| |
Collapse
|
33
|
Han ZY, Chen QW, Fu ZJ, Cheng SX, Zhang XZ. Probiotic Spore-Based Oral Drug Delivery System for Enhancing Pancreatic Cancer Chemotherapy by Gut-Pancreas-Axis-Guided Delivery. NANO LETTERS 2022; 22:8608-8617. [PMID: 36259687 DOI: 10.1021/acs.nanolett.2c03131] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The chemotherapeutic effectiveness of pancreatic ductal adenocarcinoma (PDAC) is severely hampered by insufficient intratumoral delivery of antitumor drugs. Here, we demonstrate that enhanced pancreatic cancer chemotherapy can be achieved by probiotic spore-based oral drug delivery system via gut-pancreas axis translocation. Clostridium butyricum spores resistant to harsh external stress are extracted as drug carriers, which are further covalently conjugated with gemcitabine-loaded mesoporous silicon nanoparticles (MGEM). The spore-based oral drug delivery system (SPORE-MGEM) migrates upstream into pancreatic tumors from the gut, which increases intratumoral drug accumulation by ∼3-fold compared with MGEM. In two orthotopic PDAC mice models, tumor growth is markedly suppressed by SPORE-MGEM without obvious side effects. Leveraging the biological contact of the gut-pancreas axis, this probiotic spore-based oral drug delivery system reveals a new avenue for enhancing PDAC chemotherapy.
Collapse
Affiliation(s)
- Zi-Yi Han
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Zhuang-Jiong Fu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China
- Wuhan Research Centre for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430071, People's Republic of China
| |
Collapse
|
34
|
Singh S, Singh M, Gaur S. Probiotics as multifaceted oral vaccines against colon cancer: A review. Front Immunol 2022; 13:1002674. [PMID: 36263037 PMCID: PMC9573965 DOI: 10.3389/fimmu.2022.1002674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics are known as the live microorganisms that, upon adequate administration, elicit a health beneficial response inside the host. The probiotics are known as immunomodulators and exhibit anti-tumor properties. Advanced research has explored the potential use of probiotics as the oral vaccines without the latent risks of pathogenicity. Probiotic-based oral vaccines are known to induce mucosal immunity that prevents the host from several enteric infections. Probiotic bacteria have the ability to produce metabolites in the form of anti-inflammatory cytokines, which play an important role in the prevention of carcinogenesis and in the activation of the phagocytes that eliminate the preliminary stage cancer cells. This review discusses the advantages and disadvantages of using the oral probiotic vaccines as well as the mechanism of action of probiotics in colon cancer therapy. This review also employs the use of “PROBIO” database for selecting certain probiotics with immunomodulatory properties. Furthermore, the use of several probiotic bacteria as anti-colon cancer adjuvants has also been discussed in detail. Because the current studies and trials are more focused on using the attenuated pathogens instead of using the probiotic-based vaccines, future studies must involve the advanced research in exploiting the potential of several probiotic strains as adjuvants in cancer therapies.
Collapse
Affiliation(s)
- Shubhi Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Manisha Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- *Correspondence: Smriti Gaur,
| |
Collapse
|
35
|
Huang L, Wang J, Kong L, Wang X, Li Q, Zhang L, Shi J, Duan J, Mu H. ROS-responsive hyaluronic acid hydrogel for targeted delivery of probiotics to relieve colitis. Int J Biol Macromol 2022; 222:1476-1486. [PMID: 36195227 DOI: 10.1016/j.ijbiomac.2022.09.247] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Probiotics are generally used as therapeutic intervention in inflammatory bowel disease. However, the low survival rate in harsh gastrointestinal environment and limited retention in intestine greatly restrict their health benefits. To address this problem, a ROS-responsive hydrogel based on hyaluronic acid (HA) was developed for encapsulation and targeted delivery of probiotics. The hydrogel was prepared facilely by physiological crosslink with methacrylated HA and thiolated thioketal. As a model probiotic, Lactobacillu reuteri showed a significantly increased survival rate in simulated digestive conditions after encapsulated in hydrogel. The negative properties conferred the hydrogel preferential adhesions to inflammation sites. Meanwhile, the excess reactive oxygen species (ROS) produced by inflamed colon tissues selectively cleaved thioketal linkages resulted in hydrogel degradation and local probiotics release. Furthermore, the hydrogel exerted an appropriate ROS-scavenge capacity and protected HT-29 cells from oxidative damage. Animal experiments indicated that hydrogel-encapsulated L. reuteri could remarkably alleviate the symptoms and improve the survival rate of mice with dextran sulfate sodium (DSS)-induced colitis. These results suggested that the biocompatible hydrogel may be a delivery platform to target inflamed intestines and expand the application of probiotics as pharmaceuticals.
Collapse
Affiliation(s)
- Lijie Huang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junjie Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lili Kong
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xing Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qiulei Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lingjiao Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jingru Shi
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jinyou Duan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haibo Mu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
36
|
Wang J, Guo N, Hou W, Qin H. Coating bacteria for anti-tumor therapy. Front Bioeng Biotechnol 2022; 10:1020020. [PMID: 36185433 PMCID: PMC9520470 DOI: 10.3389/fbioe.2022.1020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Therapeutic bacteria have shown great potential on anti-tumor therapy. Compared with traditional therapeutic strategy, living bacteria present unique advantages. Bacteria show high targeting and great colonization ability in tumor microenvironment with hypoxic and nutritious conditions. Bacterial-medicated antitumor therapy has been successfully applied on mouse models, but the low therapeutic effect and biosafe limit its application on clinical treatment. With the development of material science, coating living bacteria with suitable materials has received widespread attention to achieve synergetic therapy on tumor. In this review, we summarize various materials for coating living bacteria in cancer therapy and envision the opportunities and challenges of bacteria-medicated antitumor therapy.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Ning Guo
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Ning Guo, ; Weiliang Hou, ; Huanlong Qin,
| | - Weiliang Hou
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Ning Guo, ; Weiliang Hou, ; Huanlong Qin,
| | - Huanlong Qin
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Ning Guo, ; Weiliang Hou, ; Huanlong Qin,
| |
Collapse
|
37
|
Liu H, Zhang Q, Wang S, Weng W, Jing Y, Su J. Bacterial extracellular vesicles as bioactive nanocarriers for drug delivery: Advances and perspectives. Bioact Mater 2022; 14:169-181. [PMID: 35310361 PMCID: PMC8892084 DOI: 10.1016/j.bioactmat.2021.12.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Nanosized extracellular vesicles derived from bacteria contain diverse cargo and transfer intercellular bioactive molecules to cells. Due to their favorable intercellular interactions, cell membrane-derived bacterial extracellular vesicles (BEVs) have great potential to become novel drug delivery platforms. In this review, we summarize the biogenesis mechanism and compositions of various BEVs. In addition, an overview of effective isolation and purification techniques of BEVs is provided. In particular, we focus on the application of BEVs as bioactive nanocarriers for drug delivery. Finally, we summarize the advances and challenges of BEVs after providing a comprehensive discussion in each section. We believe that a deeper understanding of BEVs will open new avenues for their exploitation in drug delivery applications. Bacterial extracellular vesicles (BEVs) are excellent nanomaterials as drug delivery systems. The unique nanosized structures and biofunctions of BEVs are attractive for their use as nanomedicine platforms. BEVs have been investigated as biotherapeutics due to their loading capacity, ease of modification and industrialization. This review provides new insights of BEVs in drug delivery applications, discussing potential opportunities and challenges.
Collapse
Affiliation(s)
- Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Weizong Weng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Corresponding author.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Corresponding author.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Corresponding author. Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
38
|
Yang L, Hung LY, Zhu Y, Ding S, Margolis KG, Leong KW. Material Engineering in Gut Microbiome and Human Health. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9804014. [PMID: 35958108 PMCID: PMC9343081 DOI: 10.34133/2022/9804014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/10/2022] [Indexed: 12/11/2022]
Abstract
Tremendous progress has been made in the past decade regarding our understanding of the gut microbiome's role in human health. Currently, however, a comprehensive and focused review marrying the two distinct fields of gut microbiome and material research is lacking. To bridge the gap, the current paper discusses critical aspects of the rapidly emerging research topic of "material engineering in the gut microbiome and human health." By engaging scientists with diverse backgrounds in biomaterials, gut-microbiome axis, neuroscience, synthetic biology, tissue engineering, and biosensing in a dialogue, our goal is to accelerate the development of research tools for gut microbiome research and the development of therapeutics that target the gut microbiome. For this purpose, state-of-the-art knowledge is presented here on biomaterial technologies that facilitate the study, analysis, and manipulation of the gut microbiome, including intestinal organoids, gut-on-chip models, hydrogels for spatial mapping of gut microbiome compositions, microbiome biosensors, and oral bacteria delivery systems. In addition, a discussion is provided regarding the microbiome-gut-brain axis and the critical roles that biomaterials can play to investigate and regulate the axis. Lastly, perspectives are provided regarding future directions on how to develop and use novel biomaterials in gut microbiome research, as well as essential regulatory rules in clinical translation. In this way, we hope to inspire research into future biomaterial technologies to advance gut microbiome research and gut microbiome-based theragnostics.
Collapse
Affiliation(s)
- Letao Yang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Lin Y. Hung
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Yuefei Zhu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Suwan Ding
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University, New York, New York, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| |
Collapse
|
39
|
Rakotonirina A, Galperine T, Allémann E. Fecal microbiota transplantation: a review on current formulations in Clostridioides difficile infection and future outlooks. Expert Opin Biol Ther 2022; 22:929-944. [PMID: 35763604 DOI: 10.1080/14712598.2022.2095901] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The role of the gut microbiota in health and the pathogenesis of several diseases has been highlighted in recent years. Even though the precise mechanisms involving the microbiome in these ailments are still unclear, microbiota-modulating therapies have been developed. Fecal microbiota transplantation (FMT) has shown significant results against Clostridioides difficile infection (CDI), and its potential has been investigated for other diseases. Unfortunately, the technical aspects of the treatment make it difficult to implement. Pharmaceutical technology approaches to encapsulate microorganisms could play an important role in providing this treatment and render the treatment modalities easier to handle. AREAS COVERED After an overview of CDI, this narrative review aims to discuss the current formulations for FMT and specifically addresses the technical aspects of the treatment. This review also distinguishes itself by focusing on the hurdles and emphasizing the possible improvements using pharmaceutical technologies. EXPERT OPINION FMT is an efficient treatment for recurrent CDI. However, its standardization is overlooked. The approach of industrial and hospital preparations of FMT are different, but both show promise in their respective methodologies. Novel FMT formulations could enable further research on dysbiotic diseases in the future.
Collapse
Affiliation(s)
- Adèle Rakotonirina
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Tatiana Galperine
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland.,French Group of Faecal Microbiota Transplantation
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Miri AH, Kamankesh M, Llopis-Lorente A, Liu C, Wacker MG, Haririan I, Asadzadeh Aghdaei H, Hamblin MR, Yadegar A, Rad-Malekshahi M, Zali MR. The Potential Use of Antibiotics Against Helicobacter pylori Infection: Biopharmaceutical Implications. Front Pharmacol 2022; 13:917184. [PMID: 35833028 PMCID: PMC9271669 DOI: 10.3389/fphar.2022.917184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a notorious, recalcitrant and silent germ, which can cause a variety of debilitating stomach diseases, including gastric and duodenal ulcers and gastric cancer. This microbe predominantly colonizes the mucosal layer of the human stomach and survives in the inhospitable gastric microenvironment, by adapting to this hostile milieu. In this review, we first discuss H. pylori colonization and invasion. Thereafter, we provide a survey of current curative options based on polypharmacy, looking at pharmacokinetics, pharmacodynamics and pharmaceutical microbiology concepts, in the battle against H. pylori infection.
Collapse
Affiliation(s)
- Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, Tehran, Iran
| | - Antoni Llopis-Lorente
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, China
| | - Matthias G. Wacker
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| |
Collapse
|
41
|
Chen S, Yao W, Wang H, Wang T, Xiao X, Sun G, Yang J, Guan Y, Zhang Z, Xia Z, Li M, Tao Y, Hei Z. Injectable electrospun fiber-hydrogel composite sequentially releasing clonidine and ropivacaine for prolonged and walking regional analgesia. Am J Cancer Res 2022; 12:4904-4921. [PMID: 35836801 PMCID: PMC9274753 DOI: 10.7150/thno.74845] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023] Open
Abstract
Rationale: Peripheral nerve block is a traditional perioperative analgesic method for its precise pain control and low systemic toxicity. However, a single low dose of local anesthetic merely provides a few hours of analgesia, and high dose results in irreversible toxicity, whereas continuous infusion of anesthetics is expensive and complicated. Therefore, it is necessary to develop a long-acting and sensory-selective local anesthetic for safe perioperative analgesia. Methods: An injectable composite comprising ropivacaine-loaded poly (ε-caprolactone) electrospun fiber and clonidine-loaded F127 hydrogel (Fiber-Rop/Gel-Clo composite) was developed for long-acting and walking regional analgesia with barely one dose. The peripheral nerve blockade effect of the composite was evaluated in a rat sciatic nerve block model. Also, the biodegradability and biosafety of the composite was evaluated. Results: The preferentially released Clo from the hydrogel rapidly constricted the peripheral arterial vessels, reducing the blood absorption of Rop and thus enhancing the local Rop accumulation at the injection site. The subsequently sustainable release of Rop from the fiber, significantly prolonged the sciatic nerve block of rats. Remarkably, an amazing sensorimotor segregation effect was achieved, as the sensory blockade (32.0 ± 1.4 h) lasted significantly longer than the motor blockade (20.3 ± 0.9 h). Additionally, the Fiber-Rop/Gel-Clo composite presented good biodegradability and biosafety in vivo. Conclusions: Our designed Fiber-Rop/Gel-Clo composite with minimal invasion, prolonged synergistic analgesia, and strikingly sensorimotor segregation effect, posted a promising prospect for regional long-term walking analgesia in clinical treatment.
Collapse
Affiliation(s)
- Sufang Chen
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Weifeng Yao
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tienan Wang
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xue Xiao
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Guoliang Sun
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jing Yang
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Guan
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhen Zhang
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhengyuan Xia
- Department of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Mingqiang Li
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China.,✉ Corresponding authors: Email addresses: (M. Li), (Y. Tao), (Z. Hei)
| | - Yu Tao
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,✉ Corresponding authors: Email addresses: (M. Li), (Y. Tao), (Z. Hei)
| | - Ziqing Hei
- Department of Anesthesiology and Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,✉ Corresponding authors: Email addresses: (M. Li), (Y. Tao), (Z. Hei)
| |
Collapse
|
42
|
Abstract
With the outstanding achievement of chimeric antigen receptor (CAR)-T cell therapy in the clinic, cell-based medicines have attracted considerable attention for biomedical applications and thus generated encouraging progress. As the basic construction unit of organisms, cells harbor low immunogenicity, desirable compatibility, and a strong capability of crossing various biological barriers. However, there is still a long way to go to fix significant bottlenecks for their clinical translation, such as facile preparation, strict stability requirements, scale-up manufacturing, off-target toxicity, and affordability. The rapid development of biotechnology and engineering approaches in materials sciences has provided an ideal platform to assist cell-based therapeutics for wide application in disease treatments by overcoming these issues. Herein, we survey the most recent advances of various cells as bioactive ingredients and outline the roles of biomaterials in developing cell-based therapeutics. Besides, a perspective of cell therapies is offered with a particular focus on biomaterial-involved development of cell-based biopharmaceuticals.
Collapse
Affiliation(s)
- Yu Chen
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Samira Pal
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA. .,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.,Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
43
|
Jingushi K, Kawashima A, Saito T, Kanazawa T, Motooka D, Kimura T, Mita M, Yamamoto A, Uemura T, Yamamichi G, Okada K, Tomiyama E, Koh Y, Matsushita M, Kato T, Hatano K, Uemura M, Tsujikawa K, Wada H, Nonomura N. Circulating extracellular vesicles carrying Firmicutes reflective of the local immune status may predict clinical response to pembrolizumab in urothelial carcinoma patients. Cancer Immunol Immunother 2022; 71:2999-3011. [DOI: 10.1007/s00262-022-03213-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
|
44
|
Li F, Zhang J, Yi K, Wang H, Wei H, Chan HF, Tao Y, Li M. Delivery of Stem Cell Secretome for Therapeutic Applications. ACS APPLIED BIO MATERIALS 2022; 5:2009-2030. [PMID: 35285638 DOI: 10.1021/acsabm.1c01312] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intensive studies on stem cell therapy reveal that benefits of stem cells attribute to the paracrine effects. Hence, direct delivery of stem cell secretome to the injured site shows the comparative therapeutic efficacy of living cells while avoiding the potential limitations. However, conventional systemic administration of stem cell secretome often leads to rapid clearance in vivo. Therefore, a variety of different biomaterials are developed for sustained and controllable delivery of stem cell secretome to improve therapeutic efficiency. In this review, we first introduce current approaches for the preparation and characterization of stem cell secretome as well as strategies to improve their therapeutic efficacy and production. The up-to-date delivery platforms are also summarized, including nanoparticles, injectable hydrogels, microneedles, and scaffold patches. Meanwhile, we discuss the underlying therapeutic mechanism of stem cell secretome for the treatment of various diseases. In the end, future opportunities and challenges are proposed.
Collapse
Affiliation(s)
- Fenfang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hongyan Wei
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
45
|
Qin Y, Lao YH, Wang H, Zhang J, Yi K, Chen Z, Han J, Song W, Tao Y, Li M. Combatting Helicobacter pylori with oral nanomedicines. J Mater Chem B 2021; 9:9826-9838. [PMID: 34854456 DOI: 10.1039/d1tb02038b] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori (H. pylori) infection is considered to be the main cause of most digestive diseases,such as chronic active gastritis, gastroduodenal ulcers, or even gastric cancer. Oral medication is a transformative approach to treat H. pylori-induced infections. However, unlike intravenous administration, orally administrated drugs have to overcome various barriers before reaching the infected sites, which significantly limits the therapeutic efficacy. These challenges may be addressed by emerging nanomedicine that is equipped with nanotechnology approaches to enable efficient and effective targeted delivery of drugs. Herein, in this review, we first discuss the conventional therapy for the eradication of H. pylori. Through the introduction of the critical barriers of oral administration, the benefits of nanomedicine are highlighted. Recently-published examples of nanocarriers for combating H. pylori in terms of design, preparation, and antimicrobial mechanisms are then presented, followed by our perspective on potential future research directions of oral nanomedicines.
Collapse
Affiliation(s)
- Yuan Qin
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Jiabin Zhang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Zhuanggui Chen
- Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jing Han
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
46
|
Su R, Zhang Y, Zhang J, Wang H, Luo Y, Chan HF, Tao Y, Chen Z, Li M. Nanomedicine to advance the treatment of bacteria-induced acute lung injury. J Mater Chem B 2021; 9:9100-9115. [PMID: 34672317 DOI: 10.1039/d1tb01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacteria-induced acute lung injury (ALI) is associated with a high mortality rate due to the lack of an effective treatment. Patients often rely on supportive care such as low tidal volume ventilation to alleviate the symptoms. Nanomedicine has recently received much attention owing to its premium benefits of delivering drugs in a sustainable and controllable manner while minimizing the potential side effects. It can effectively improve the prognosis of bacteria-induced ALI through targeted delivery of drugs, regulation of multiple inflammatory pathways, and combating antibiotic resistance. Hence, in this review, we first discuss the pathogenesis of ALI and its potential therapeutics. In particular, the state-of-the-art nanomedicines for the treatment of bacteria-induced ALI are highlighted, including their administration routes, in vivo distribution, and clearance. Furthermore, the available bacteria-induced ALI animal models are also summarized. In the end, future perspectives of nanomedicine for ALI treatment are proposed.
Collapse
Affiliation(s)
- Ruonan Su
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca 14853, USA
| | - Jiabin Zhang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haixia Wang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhuanggui Chen
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
47
|
Filip R, Anchidin-Norocel L, Gheorghita R, Savage WK, Dimian M. Changes in Dietary Patterns and Clinical Health Outcomes in Different Countries during the SARS-CoV-2 Pandemic. Nutrients 2021; 13:3612. [PMID: 34684615 PMCID: PMC8539259 DOI: 10.3390/nu13103612] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to an excess in community mortality across the globe. We review recent evidence on the clinical pathology of COVID-19, comorbidity factors, immune response to SARS-CoV-2 infection, and factors influencing infection outcomes. The latter specifically includes diet and lifestyle factors during pandemic restrictions. We also cover the possibility of SARS-CoV-2 transmission through food products and the food chain, as well as virus persistence on different surfaces and in different environmental conditions, which were major public concerns during the initial days of the pandemic, but have since waned in public attention. We discuss useful measures to avoid the risk of SARS-CoV-2 spread through food, and approaches that may reduce the risk of contamination with the highly contagious virus. While hygienic protocols are required in food supply sectors, cleaning, disinfection, avoidance of cross-contamination across food categories, and foodstuffs at different stages of the manufacturing process are still particularly relevant because the virus persists at length on inert materials such as food packaging. Moreover, personal hygiene (frequent washing and disinfection), wearing gloves, and proper use of masks, clothes, and footwear dedicated to maintaining hygiene, provide on-site protections for food sector employees as well as supply chain intermediates and consumers. Finally, we emphasize the importance of following a healthy diet and maintaining a lifestyle that promotes physical well-being and supports healthy immune system function, especially when government movement restrictions ("lockdowns") are implemented.
Collapse
Affiliation(s)
- Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.F.); (R.G.); (W.K.S.)
- Regional County Emergency Hospital, 720224 Suceava, Romania
| | - Liliana Anchidin-Norocel
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.F.); (R.G.); (W.K.S.)
| | - Roxana Gheorghita
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.F.); (R.G.); (W.K.S.)
- Integrated Center for Research, Development and Innovation in Advanced Materials, Nanotechnologies, and Distributed Systems for Fabrication and Control, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
| | - Wesley K. Savage
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, 720229 Suceava, Romania; (R.F.); (R.G.); (W.K.S.)
- Integrated Center for Research, Development and Innovation in Advanced Materials, Nanotechnologies, and Distributed Systems for Fabrication and Control, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
| | - Mihai Dimian
- Integrated Center for Research, Development and Innovation in Advanced Materials, Nanotechnologies, and Distributed Systems for Fabrication and Control, Stefan cel Mare University of Suceava, 720229 Suceava, Romania;
- Department of Computers, Electronics and Automation, Stefan cel Mare University of Suceava, 720229 Suceava, Romania
| |
Collapse
|
48
|
Qiu K, Anselmo AC. Enhanced Storage of Anaerobic Bacteria through Polymeric Encapsulation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46282-46290. [PMID: 34558893 DOI: 10.1021/acsami.1c11785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Live microbes such as lactobacilli have long been used as probiotic supplements and, more recently, have been explored as live biotherapeutic products with the potential to treat a range of conditions. Among these microbes is a category of anaerobes that possess therapeutic potential while exhibiting unique oxygen sensitivity and thus requiring careful considerations in the formulation and storage processes. Existing microbial formulation development has focused on facultative anaerobes with natural oxygen tolerance; a few strategies have been reported for anaerobes with demonstrated oxygen intolerance, warranting novel approaches toward addressing the challenges for these oxygen-sensitive anaerobes. Here, we develop a polymeric encapsulation system for the formulation and storage of Bifidobacterium adolescentis (B. adolescentis), a model anaerobe that loses viability in aerobic incubation at 37 °C within 1 day. We discover that this strain remains viable under aerobic conditions for 14 days at 4 °C, enabling formulation development such as solution casting and air drying in an aerobic environment. Next, through a systematic selection of polymer encapsulants and excipients, we show that encapsulation with poly(vinyl alcohol) (PVA) acts as an oxygen barrier and facilitates long-term storage of B. adolescentis, which is partially attributed to reduced generation of reactive oxygen species. Lastly, PVA-based formulations can produce oral capsule-loaded films and edible gummy bears, demonstrating its compatibility with both pharmaceutical and food dosage forms.
Collapse
Affiliation(s)
- Kunyu Qiu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Aaron C Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
49
|
Bacteria-Based Microdevices for the Oral Delivery of Macromolecules. Pharmaceutics 2021; 13:pharmaceutics13101610. [PMID: 34683903 PMCID: PMC8537518 DOI: 10.3390/pharmaceutics13101610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The oral delivery of macromolecules is quite challenging due to environmental insults and biological barriers encountered along the gastrointestinal (GI) tract. Benefiting from their living characteristics, diverse bacterial species have been engineered as intelligent platforms to deliver various therapeutics. To tackle difficulties in oral delivery, innovative bacteria-based microdevices have been developed by virtue of advancements in synthetic biology and nanotechnology, with aims to overcome the instability and short half-life of macromolecules in the GI tract. In this review, we summarize the main classes of macromolecules that are produced and delivered through the oral ingestion of bacteria and bacterial derivatives. Furtherly, we discuss the engineering strategies and biomedical applications of these living microdevices in disease diagnosis, bioimaging, and treatment. Finally, we highlight the advantages as well as the limitations of these engineered bacteria used as platforms for the oral delivery of macromolecules and also propose their potential for clinical translation. The results summarized in this review article would contribute to the invention of next-generation bacteria-based systems for the oral delivery of macromolecules.
Collapse
|
50
|
Qiu K, Huang Y, Anselmo AC. Polymer and Crosslinker Content Influences Performance of Encapsulated Live Biotherapeutic Products. Cell Mol Bioeng 2021; 14:487-499. [PMID: 34777606 PMCID: PMC8548438 DOI: 10.1007/s12195-021-00674-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/27/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Live biotherapeutic products (LBPs), or therapeutic microbes, are an emerging therapeutic modality for prevention and treatment of gastrointestinal diseases. Since LBPs are living, they are uniquely sensitive to external stresses (e.g., oxygen, acid) encountered during manufacturing, storage, and delivery. Here, we systematically evaluate how polymer and crosslinker concentration affects the performance of an encapsulated LBP toward developing a comprehensive framework for the characterization and optimization of LBP delivery systems. METHODS We encapsulate a model LBP, Lactobacillus casei ATCC 393, in calcium chloride (CaCl2)-crosslinked alginate beads, and evaluate how alginate and CaCl2 concentrations influence LBP formulation performance, including: (i) encapsulation efficiency, (ii) shrinkage upon drying, (iii) survival upon lyophilization, (iv) acid resistance, (v) release, and (vi) metabolite secretion. Approaches from microbiology (e.g., colony forming unit enumeration), materials science (e.g., scanning electron microscopy), and pharmaceutical sciences (e.g., release assays) are employed. RESULTS LBP-encapsulating alginate beads were systematically evaluated as a function of alginate and CaCl2 concentrations. Specifically: (i) encapsulation efficiency of all formulations was >50%, (ii) all alginate beads shrunk (after lyophilization) and recovered (after rehydration) similarly, (iii) at 10% alginate concentration, lower CaCl2 concentration decreased survival upon lyophilization, (iv) 10% alginate improved acid resistance, (v) sustained release was enabled by increasing alginate and CaCl2 concentrations, and (vi) encapsulation did not impair secretion of l-lactate as compared to free LBP. CONCLUSIONS This research demonstrates that polymer content and crosslinking extent modulate the performance of polymer-based LBP delivery systems, motivating research into the optimization of material properties for LBP delivery systems.
Collapse
Affiliation(s)
- Kunyu Qiu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 United States
| | - Yirui Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 United States
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 United States
| |
Collapse
|