1
|
Zhang Y, He J, Zeng H, Xu D, Li W, Wang Y. Advances in prebiotic carbohydrate-based targeted delivery: Overcoming gastrointestinal challenges for bioactive ingredients. Food Chem 2025; 466:142210. [PMID: 39615354 DOI: 10.1016/j.foodchem.2024.142210] [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: 07/24/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024]
Abstract
Natural bioactive ingredients face challenges in extensive application owing to low oral bioavailability. This can be improved by overcoming gastrointestinal barriers and facilitating targeted release through delivery strategies. This study provides a comprehensive review of targeted delivery systems using prebiotic carbohydrate matrices, focusing on structures, release mechanisms and applications. The bioactive ingredients can be encapsulated into nanohydrogels, nanoparticles, nanoemulsions, micro/nanocapsules and nanofibres to achieve controlled/targeted delivery to predetermined locations via interactions with pH, mucus, microbiome, enzymes and other factors in the colon. In particular, the prebiotic function of carbohydrates is generated by colonic microbiota degradation and fermentation, producing beneficial postbiotics through multiple metabolic pathways. This study provides certain insights into the in-depth development and application of prebiotic carbohydrate-based targeted delivery systems in the fields of food and health.
Collapse
Affiliation(s)
- Yunzhen Zhang
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, 8 West Guochuang Road, Hohhot 010110, Inner Mongolia, PR China
| | - Hong Zeng
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Duoxia Xu
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Wenlu Li
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China.
| | - Yanbo Wang
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China.
| |
Collapse
|
2
|
Guo J, Qiu Y, Zhang J, Xue C, Zhu J. A review on polysaccharide-based delivery systems for edible bioactives: pH responsive, controlled release, and emerging applications. Int J Biol Macromol 2024; 291:139178. [PMID: 39730044 DOI: 10.1016/j.ijbiomac.2024.139178] [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: 09/01/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
pH changes occur during bodily lesions, presenting an opportunity for leveraging pH-responsive delivery systems as signals for a targeted response. This review explores the design and application of pH-responsive delivery systems based on natural polysaccharides for the controlled release of bioactives. The article examines the development of diverse delivery carriers, including nanoparticles, nanofibers, nanogels, core-shell carriers, hydrogels, emulsions as well as liposomes and their capacity to respond to pH variations, enabling the precise and targeted delivery of bioactives within the human body. These polysaccharide-based delivery systems can be made pH-responsive by modulating the charge of polybasic or polyacidic polysaccharides, inducing swelling of the carrier and subsequent release of the encapsulated bioactives. These pH-responsive systems show promise in stabilizing under acidic conditions for enhanced retention in the stomach during oral delivery while also enabling targeted release at low pH sites such as tumors and wounds, thereby accelerating wound healing and aiding in cancer therapy and inflammation treatment. pH can co-respond with a variety of stimuli, including temperature, enzymes and reactive oxygen species, enabling more precise responses to the microenvironment for targeted delivery. It provides solid theoretical foundations for the advancement of personalized nutrition and therapeutics through controlled and responsive release technologies.
Collapse
Affiliation(s)
- Jiaxin Guo
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yang Qiu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Chenxu Xue
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Zhu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
3
|
Liang W, Zhang W, Tian J, Zhang X, Lv X, Qu A, Chen J, Wu Z. Advances in carbohydrate-based nanoparticles for targeted therapy of inflammatory bowel diseases: A review. Int J Biol Macromol 2024; 281:136392. [PMID: 39423983 DOI: 10.1016/j.ijbiomac.2024.136392] [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: 05/18/2024] [Revised: 09/13/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
The incidence of inflammatory bowel disease (IBD), a chronic gastrointestinal disorder, is rapidly increasing worldwide. Unfortunately, the current therapies for IBD are often hindered by premature drug release and undesirable side effects. With the advancement of nanotechnology, the innovative targeted nanotherapeutics are explored to ensure the accurate delivery of drugs to specific sites in the colon, thereby reducing side effects and improving the efficacy of oral administration. The emphasis of this review is to summarize the potential pathogenesis of IBD and highlight recent breakthroughs in carbohydrate-based nanoparticles for IBD treatment, including their construction, release mechanism, potential targeting ability, and their therapeutic efficacy. Specifically, we summarize the latest knowledge regarding environmental-responsive nano-systems and active targeted nanoparticles. The environmental-responsive drug delivery systems crafted with carbohydrates or other biological macromolecules like chitosan and sodium alginate, exhibit a remarkable capacity to enhance the accumulation of therapeutic drugs in the inflamed regions of the digestive tract. Active targeting strategies improve the specificity and accuracy of oral drug delivery to the colon by modifying carbohydrates such as hyaluronic acid and mannose onto nanocarriers. Finally, we discuss the challenges and provide insight into the future perspectives of colon-targeted delivery systems for IBD treatment.
Collapse
Affiliation(s)
- Wenjing Liang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Wen Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China.
| | - Jiayi Tian
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Xinping Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Xinyi Lv
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Ao Qu
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Jinyu Chen
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China
| | - Zijian Wu
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China.
| |
Collapse
|
4
|
Long J, Liang X, Ao Z, Tang X, Li C, Yan K, Yu X, Wan Y, Li Y, Li C, Zhou M. Stimulus-responsive drug delivery nanoplatforms for inflammatory bowel disease therapy. Acta Biomater 2024; 188:27-47. [PMID: 39265673 DOI: 10.1016/j.actbio.2024.09.007] [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: 05/22/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
Inflammatory bowel disease (IBD) manifests as inflammation in the colon, rectum, and ileum, presenting a global health concern with increasing prevalence. Therefore, effective anti-inflammatory therapy stands as a promising strategy for the prevention and management of IBD. However, conventional nano drug delivery systems (NDDSs) for IBD face many challenges in targeting the intestine, such as physiological and pathological barriers, genetic variants, disease severity, and nutritional status, which often result in nonspecific tissue distribution and uncontrolled drug release. To address these limitations, stimulus-responsive NDDSs have received considerable attention in recent years due to their advantages in providing controlled release and enhanced targeting. This review provides an overview of the pathophysiological mechanisms underlying IBD and summarizes recent advancements in microenvironmental stimulus-responsive nanocarriers for IBD therapy. These carriers utilize physicochemical stimuli such as pH, reactive oxygen species, enzymes, and redox substances to deliver drugs for IBD treatment. Additionally, pivotal challenges in the future development and clinical translation of stimulus-responsive NDDSs are emphasized. By offering insights into the development and optimization of stimulus-responsive drug delivery nanoplatforms, this review aims to facilitate their application in treating IBD. STATEMENT OF SIGNIFICANCE: This review highlights recent advancements in stimulus-responsive nano drug delivery systems (NDDSs) for the treatment of inflammatory bowel disease (IBD). These innovative nanoplatforms respond to specific environmental triggers, such as pH reactive oxygen species, enzymes, and redox substances, to release drugs directly at the inflammation site. By summarizing the latest research, our work underscores the potential of these technologies to improve drug targeting and efficacy, offering new directions for IBD therapy. This review is significant as it provides a comprehensive overview for researchers and clinicians, facilitating the development of more effective treatments for IBD and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jiang Long
- Department of Cardiology, Xuyong County People's Hospital, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao Tang
- College of Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kexin Yan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Science and Technology Department, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
5
|
Li H, Cheng Y, Cui L, Yang Z, Wang J, Zhang Z, Chen K, Zhao C, He N, Li S. Combining Gut Microbiota Modulation and Enzymatic-Triggered Colonic Delivery by Prebiotic Nanoparticles Improves Mouse Colitis Therapy. Biomater Res 2024; 28:0062. [PMID: 39140035 PMCID: PMC11321063 DOI: 10.34133/bmr.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 07/02/2024] [Indexed: 08/15/2024] Open
Abstract
The efficacy of ulcerative colitis (UC) therapy is closely connected to the composition of gut microbiota in the gastrointestinal tract. Prebiotic-based nanoparticles (NPs) provide a more precise approach to alleviate UC via modulating gut microbiota dysbiosis. The present study develops an efficient prebiotic-based colon-targeted drug delivery system (PCDDS) by using prebiotic pectin (Pcn) and chitosan (Csn) polysaccharides as a prebiotic shell, with the anti-inflammatory drug sulfasalazine (SAS) loaded into a poly(lactic-co-glycolic acid) (PLGA) core to construct SAS@PLGA-Csn-Pcn NPs. Then, we examine its characterization, cellular uptake, and in vivo therapeutic efficacy. The results of our study indicate that the Pcn/Csn shell confers efficient pH-sensitivity properties. The gut microbiota-secreted pectinase serves as the trigger agent for Pcn/Csn shell degradation, and the resulting Pcn oligosaccharides possess a substantial prebiotic property. Meanwhile, the formed PCDDSs exhibit robust biodistribution and accumulation in the colon tissue, rapid cellular uptake, efficient in vivo therapeutic efficacy, and modulation of gut microbiota dysbiosis in a mouse colitis model. Collectively, our synthetic PCDDSs demonstrate a promising and synergistic strategy for UC therapy.
Collapse
Affiliation(s)
- Hui Li
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Yu Cheng
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Luwen Cui
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Jingyi Wang
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Zixuan Zhang
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Kaiwei Chen
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Cheng Zhao
- Department of Abdominal Ultrasound,
The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College,
Qingdao University, Qingdao, China
- Department of Abdominal Ultrasound,
The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Shi J, Zhou J, Liu B, Lin K, Xie X, Han X, Sheng Y, Liu Y, He C, Zhou Y, Zhu N, Yang Q, Luo R, Li Y. Enzyme/ROS dual-sensitive nanoplatform with on-demand Celastrol release capacity for enhanced ulcerative colitis therapy by ROS scavenging, microbiota rebalancing, inflammation alleviating. J Nanobiotechnology 2024; 22:437. [PMID: 39061092 PMCID: PMC11282782 DOI: 10.1186/s12951-024-02725-9] [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: 03/24/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND The oral administration of drugs for treating ulcerative colitis (UC) is hindered by several factors, including inadequate gastrointestinal stability, insufficient accumulation in colonic lesions, and uncontrolled drug release. METHODS A multiple sensitive nano-delivery system comprising β-cyclodextrin (CD) and 4-(hydroxymethyl)phenylboronic acid (PAPE) with enzyme/reactive oxygen species (ROS) sensitivity was developed to load celastrol (Cel) as a comprehensive treatment for UC. RESULTS Owing to the positive charge in the site of inflamed colonic mucosa, the negatively charged nanomedicine (Cel/NPs) could efficiently accumulate. Expectedly, Cel/NPs showed excellent localization ability to colon in vitro and in vivo tests. The elevated concentration of ROS and intestinal enzymes in the colon microenvironment quickly break the CD, resulting in Cel release partially to rebalance microbiota and recover the intestinal barrier. The accompanying cellular internalization of residual Cel/NPs, along with the high concentration of cellular ROS to trigger Cel burst release, could decrease the expression of inflammatory cytokines, inhibit colonic cell apoptosis, promote the macrophage polarization, scavenge ROS, and regulate the TLR4/NF-κB signaling pathway, which certified that Cel/NPs possessed a notably anti-UC therapy outcome. CONCLUSIONS We provide a promising strategy for addressing UC symptoms via an enzyme/ROS-sensitive oral platform capable of releasing drugs on demand.
Collapse
Affiliation(s)
- Jinfeng Shi
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Jiahui Zhou
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Bo Liu
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Kezhou Lin
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Xingliang Xie
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Xue Han
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Yanmei Sheng
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Yihan Liu
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Congjian He
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Yujin Zhou
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Nan Zhu
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Qian Yang
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China
| | - Ruifeng Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China.
| | - Yi Li
- College of Pharmacy, Chengdu Medical College, No.783 Xindu Avenue, Xindu District, Chengdu, 610500, China.
| |
Collapse
|
7
|
Li W, Zhan M, Wen Y, Chen Y, Zhang Z, Wang S, Tian D, Tian S. Recent Progress of Oral Functional Nanomaterials for Intestinal Microbiota Regulation. Pharmaceutics 2024; 16:921. [PMID: 39065618 PMCID: PMC11280463 DOI: 10.3390/pharmaceutics16070921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The gut microbiota is closely associated with human health, and alterations in gut microbiota can influence various physiological and pathological activities in the human body. Therefore, microbiota regulation has become an important strategy in current disease treatment, albeit facing numerous challenges. Nanomaterials, owing to their excellent protective properties, drug release capabilities, targeting abilities, and good biocompatibility, have been widely developed and utilized in pharmaceuticals and dietary fields. In recent years, significant progress has been made in research on utilizing nanomaterials to assist in regulating gut microbiota for disease intervention. This review explores the latest advancements in the application of nanomaterials for microbiota regulation and offers insights into the future development of nanomaterials in modulating gut microbiota.
Collapse
Affiliation(s)
- Wanneng Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Minle Zhan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Yue Wen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Zhongchao Zhang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (W.L.); (Y.W.); (Y.C.); (Z.Z.); (S.W.)
| | - Sidan Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China;
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
8
|
Gazzi R, Gelli R, Aleandri S, Carone M, Luciani P. Bioinspired and bioderived nanomedicine for inflammatory bowel disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1986. [PMID: 39140489 DOI: 10.1002/wnan.1986] [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: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 08/15/2024]
Abstract
Due to its chronic nature and complex pathophysiology, inflammatory bowel disease (IBD) poses significant challenges for treatment. The long-term therapies for patients, often diagnosed between the ages of 20 and 40, call for innovative strategies to target inflammation, minimize systemic drug exposure, and improve patients' therapeutic outcomes. Among the plethora of strategies currently pursued, bioinspired and bioderived nano-based formulations have garnered interest for their safety and versatility in the management of IBD. Bioinspired nanomedicine can host and deliver not only small drug molecules but also biotherapeutics, be made gastroresistant and mucoadhesive or mucopenetrating and, for these reasons, are largely investigated for oral administration, while surprisingly less for rectal delivery, recommended first-line treatment approach for several IBD patients. The use of bioderived nanocarriers, mostly extracellular vesicles (EVs), endowed with unique homing abilities, is still in its infancy with respect to the arsenal of nanomedicine under investigation for IBD treatment. An emerging source of EVs suited for oral administration is ingesta, that is, plants or milk, thanks to their remarkable ability to resist the harsh environment of the upper gastrointestinal tract. Inspired by the unparalleled properties of natural biomaterials, sophisticated avenues for enhancing therapeutic efficacy and advancing precision medicine approaches in IBD care are taking shape, although bottlenecks arising either from the complexity of the nanomedicine designed or from the lack of a clear regulatory pathway still hinder a smooth and efficient translation to the clinics. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Rafaela Gazzi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Rita Gelli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Florence, Italy
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Marianna Carone
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Chen Y, Feng J, Chen Y, Xia C, Yao M, Ding W, Li X, Fu X, Zheng S, Ma Y, Zou J, Lan M, Gao F. ROS-responsive nano-medicine for navigating autophagy to enhance targeted therapy of inflammatory bowel disease. Int J Pharm 2024; 659:124117. [PMID: 38615805 DOI: 10.1016/j.ijpharm.2024.124117] [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: 02/08/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disorder characterized by immune dysregulation and intestinal inflammation. Rapamycin (Ra), an mTORC1 pathway inhibitor, has shown promise for autophagy induction in IBD therapy but is associated with off-target effects and toxicity. To address these issues, we developed an oral liposome responsive to reactive oxygen species (ROS) using lipids and amphiphilic materials. We combined ketone thiol (TK) for ROS responsive and hyaluronic acid (HA) with high affinity for CD44 receptors to prepare rapamycin-loaded nanoparticle (Ra@TH). Owing to its ROS responsive characteristic, Ra@TH can achieve inflammatory colonic targeting. Additionally, Ra@TH can induce autophagy by inhibiting the mTORC1 pathway, leading to the clearance of damaged organelles, pathogenic microorganisms and oxidative stress products. Simultaneously, it also collaboratively inhibits the NF-κB pathway suppressed by the removal of ROS resulting from TK cleavage, thereby mediating the expression of inflammatory factors. Furthermore, Ra@TH enhances the expression of typical tight junction proteins, synergistically restoring intestinal barrier function. Our research not only expands the understanding of autophagy in IBD treatment but also introduces a promising therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Juewen Feng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yang Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Min Yao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiang Li
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shulei Zheng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yin Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
10
|
Wang N, Chen L, Huang W, Gao Z, Jin M. Current Advances of Nanomaterial-Based Oral Drug Delivery for Colorectal Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:557. [PMID: 38607092 PMCID: PMC11013305 DOI: 10.3390/nano14070557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024]
Abstract
Colorectal cancer (CRC) is a common malignant tumor, and traditional treatments include surgical resection and radiotherapy. However, local recurrence, distal metastasis, and intestinal obstruction are significant problems. Oral nano-formulation is a promising treatment strategy for CRC. This study introduces physiological and environmental factors, the main challenges of CRC treatment, and the need for a novel oral colon-targeted drug delivery system (OCDDS). This study reviews the research progress of controlled-release, responsive, magnetic, targeted, and other oral nano-formulations in the direction of CRC treatment, in addition to the advantages of oral colon-targeted nano-formulations and concerns about the oral delivery of related therapeutic agents to inspire related research.
Collapse
Affiliation(s)
- Nuoya Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Yanbian University, Yanji 133000, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
11
|
Chauhan S, Harwansh RK. Recent advances in nanocarrier systems for ulcerative colitis: A new era of targeted therapy and biomarker integration. J Drug Deliv Sci Technol 2024; 93:105466. [DOI: 10.1016/j.jddst.2024.105466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Hu S, Zhao R, Xu Y, Gu Z, Zhu B, Hu J. Orally-administered nanomedicine systems targeting colon inflammation for the treatment of inflammatory bowel disease: latest advances. J Mater Chem B 2023; 12:13-38. [PMID: 38018424 DOI: 10.1039/d3tb02302h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and idiopathic condition that results in inflammation of the gastrointestinal tract, leading to conditions such as ulcerative colitis and Crohn's disease. Commonly used treatments for IBD include anti-inflammatory drugs, immunosuppressants, and antibiotics. Fecal microbiota transplantation is also being explored as a potential treatment method; however, these drugs may lead to systemic side effects. Oral administration is preferred for IBD treatment, but accurately locating the inflamed area in the colon is challenging due to multiple physiological barriers. Nanoparticle drug delivery systems possess unique physicochemical properties that enable precise delivery to the target site for IBD treatment, exploiting the increased permeability and retention effect of inflamed intestines. The first part of this review comprehensively introduces the pathophysiological environment of IBD, covering the gastrointestinal pH, various enzymes in the pathway, transport time, intestinal mucus, intestinal epithelium, intestinal immune cells, and intestinal microbiota. The second part focuses on the latest advances in the mechanism and strategies of targeted delivery using oral nanoparticle drug delivery systems for colitis-related fields. Finally, we present challenges and potential directions for future IBD treatment with the assistance of nanotechnology.
Collapse
Affiliation(s)
- Shumeng Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
| | - Runan Zhao
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Xu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Zelin Gu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
| | - Beiwei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Jiangning Hu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| |
Collapse
|
13
|
Chen M, Lan H, Jin K, Chen Y. Responsive nanosystems for targeted therapy of ulcerative colitis: Current practices and future perspectives. Drug Deliv 2023; 30:2219427. [PMID: 37288799 PMCID: PMC10405869 DOI: 10.1080/10717544.2023.2219427] [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: 01/30/2023] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
The pharmacological approach to treating gastrointestinal diseases is suffering from various challenges. Among such gastrointestinal diseases, ulcerative colitis manifests inflammation at the colon site specifically. Patients suffering from ulcerative colitis notably exhibit thin mucus layers that offer increased permeability for the attacking pathogens. In the majority of ulcerative colitis patients, the conventional treatment options fail in controlling the symptoms of the disease leading to distressing effects on the quality of life. Such a scenario is due to the failure of conventional therapies to target the loaded moiety into specific diseased sites in the colon. Targeted carriers are needed to address this issue and enhance the drug effects. Conventional nanocarriers are mostly readily cleared and have nonspecific targeting. To accumulate the desired concentration of the therapeutic candidates at the inflamed area of the colon, smart nanomaterials with responsive nature have been explored recently that include pH responsive, reactive oxygen species responsive (ROS), enzyme responsive and thermo - responsive smart nanocarrier systems. The formulation of such responsive smart nanocarriers from nanotechnology scaffolds has resulted in the selective release of therapeutic drugs, avoiding systemic absorption and limiting the undesired delivery of targeting drugs into healthy tissues. Recent advancements in the field of responsive nanocarrier systems have resulted in the fabrication of multi-responsive systems i.e. dual responsive nanocarriers and derivitization that has increased the biological tissues and smart nanocarrier's interaction. In addition, it has also led to efficient targeting and significant cellular uptake of the therapeutic moieties. Herein, we have highlighted the latest status of the responsive nanocarrier drug delivery system, its applications for on-demand delivery of drug candidates for ulcerative colitis, and the prospects are underpinned.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
14
|
Zhao S, Zhao Y, Yang X, Zhao T. Recent research advances on oral colon-specific delivery system of nature bioactive components: A review. Food Res Int 2023; 173:113403. [PMID: 37803751 DOI: 10.1016/j.foodres.2023.113403] [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: 05/07/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 10/08/2023]
Abstract
Oral colon-specific delivery system (OCDS) is a targeted approach that aims to directly deliver bioactive compounds directly to the colon following oral administration, thereby enhancing the colonic release of bioactive substances and minimizing adverse reactions. The effectiveness of bioactive substances in the colon hinges on the degree of release, which are affected by various factors including pH, mucosal barrier, delivery time and so on. Therefore, this review provides a comprehensive overview of the key factors affecting oral colon-specific release of bioactive components firstly. Considering the oral safety, this review then mainly focuses on the types of carriers with edible OCDS and preparation strategies for OCDS. Finally, several preparation strategies for loading typical natural bioactive ingredients into oral safe OCDS are reviewed, along with future development prospects.
Collapse
Affiliation(s)
- Shuang Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Tong Zhao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
15
|
Swastha D, Varsha N, Aravind S, Samyuktha KB, Yokesh MM, Balde A, Ayilya BL, Benjakul S, Kim SK, Nazeer RA. Alginate-based drug carrier systems to target inflammatory bowel disease: A review. Int J Biol Macromol 2023:125472. [PMID: 37336375 DOI: 10.1016/j.ijbiomac.2023.125472] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disorder that affects the gastrointestinal tract. IBD has become an increasingly common condition in both developed and developing nations over the last few decades, owing to a variety of factors like a rising population and diets packed with processed and junk foods. While the root pathophysiology of IBD is unknown, treatments are focused on medications aimed to mitigate symptoms. Alginate (AG), a marine-derived polysaccharide, is extensively studied for its biocompatibility, pH sensitivity, and crosslinking nature. This polymer is thoroughly researched in drug delivery systems for IBD treatment, as it is naturally available, non-toxic, cost effective, and can be easily and safely cross-linked with other polymers to form an interconnected network, which helps in controlling the release of drugs over an extended period. There are various types of drug delivery systems developed from AG to deliver therapeutic agents; among them, nanotechnology-based systems and hydrogels are popular due to their ability to facilitate targeted drug delivery, reduce dosage, and increase the therapeutic efficiency. AG-based carrier systems are not only used for the sustained release of drug, but also used in the delivery of siRNA, interleukins, and stem cells for site directed drug delivery and tissue regenerating ability respectively. This review is focussed on pathogenesis and currently studied medications for IBD, AG-based drug delivery systems and their properties for the alleviation of IBD. Moreover, future challenges are also be discoursed to improve the research of AG in the field of biopharmaceuticals and drug delivery.
Collapse
Affiliation(s)
- Dinakar Swastha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Nambolan Varsha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Suresh Aravind
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Kavassery Balasubramanian Samyuktha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Muruganandam Mohaneswari Yokesh
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Bakthavatchalam Loganathan Ayilya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Soottawat Benjakul
- Department of Food Technology, Faculty of Agro-Industry, Prince of Songkhla University, 90112 Hat Yai, Songkhla, Thailand
| | - Se-Kwon Kim
- Department of Marine Science and Convergence Engineering, Hanyang University, Ansan, 11558, Gyeonggi-do, South Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India.
| |
Collapse
|
16
|
Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: Key to more efficient oral drug delivery systems. Adv Colloid Interface Sci 2023; 313:102848. [PMID: 36780780 DOI: 10.1016/j.cis.2023.102848] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As nanocarriers (NCs) can improve the solubility of drugs, prevent their degradation by gastrointestinal (GI) enzymes and promote their transport across the mucus gel layer and absorption membrane, the oral bioavailability of these drugs can be substantially enhanced. All these properties of NCs including self-emulsifying drug delivery systems (SEDDS), solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), liposomes, polymeric nanoparticles, inorganic nanoparticles and polymeric micelles depend mainly on their surface chemistry. In particular, interaction with food, digestive enzymes, bile salts and electrolytes, diffusion behaviour across the mucus gel layer and fate on the absorption membrane are determined by their surface. Bioinert surfaces limiting interactions with gastrointestinal fluid and content as well as with mucus, adhesive surfaces providing an intimate contact with the GI mucosa and absorption enhancing surfaces can be designed. Furthermore, charge converting surfaces shifting their zeta potential from negative to positive directly at the absorption membrane and surfaces providing a targeted drug release are advantageous. In addition to these passive surfaces, even active surfaces cleaving mucus glycoproteins on their way through the mucus gel layer can be created. Within this review, we provide an overview on these different surfaces and discuss their impact on the performance of NCs in the GI tract.
Collapse
Affiliation(s)
- Helen Spleis
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Matthias Sandmeier
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Victor Claus
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria.
| |
Collapse
|
17
|
Liu D, Wei M, Yan W, Xie H, Sun Y, Yuan B, Jin Y. Potential applications of drug delivery technologies against radiation enteritis. Expert Opin Drug Deliv 2023; 20:435-455. [PMID: 36809906 DOI: 10.1080/17425247.2023.2183948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
INTRODUCTION The incidence of abdominal tumors, such as colorectal and prostate cancers, continually increases. Radiation therapy is widely applied in the clinical treatment of patients with abdominal/pelvic cancers, but it often unfortunately causes radiation enteritis (RE) involving the intestine, colon, and rectum. However, there is a lack of suitable treatment options for effective prevention and treatment of RE. AREAS COVERED Conventional clinical drugs for preventing and treating RE are usually applied by enemas and oral administration. Innovative gut-targeted drug delivery systems including hydrogels, microspheres, and nanoparticles are proposed to improve the prevention and curation of RE. EXPERT OPINION The prevention and treatment of RE have not attracted sufficient attention in the clinical practice, especially compared to the treatment of tumors, although RE takes patients great pains. Drug delivery to the pathological sites of RE is a huge challenge. The short retention and weak targeting of conventional drug delivery systems affect the therapeutic efficiency of anti-RE drugs. Novel drug delivery systems including hydrogels, microspheres, and nanoparticles can allow drugs long-term retention in the gut and targeting the inflammation sites to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Xie
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingbao Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
18
|
Laurindo LF, de Carvalho GM, de Oliveira Zanuso B, Figueira ME, Direito R, de Alvares Goulart R, Buglio DS, Barbalho SM. Curcumin-Based Nanomedicines in the Treatment of Inflammatory and Immunomodulated Diseases: An Evidence-Based Comprehensive Review. Pharmaceutics 2023; 15:pharmaceutics15010229. [PMID: 36678859 PMCID: PMC9861982 DOI: 10.3390/pharmaceutics15010229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Curcumin (CUR) is a polyphenol extracted from the rhizome of Curcuma longa that possesses potent anti-inflammatory and antioxidant potential. Despite CUR's numerous beneficial effects on human health, it has limitations, such as poor absorption. Nano-based drug delivery systems have recently been applied to improve CUR's solubility and bioavailability and potentialize its health effects. This review investigated the effects of different CUR-based nanomedicines on inflammatory and immunomodulated diseases. PUBMED, EMBASE, COCHRANE, and GOOGLE SCHOLAR databases were searched, and the Scale for Assessment of Narrative Review Articles (SANRA) was used for quality assessment and PRISMA guidelines. Overall, 66 studies were included comprising atherosclerosis, rheumatoid arthritis (RA), Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), Huntington's disease (HD), inflammatory bowel diseases (IBD), psoriasis, liver fibrosis, epilepsy, and COVID-19. The available scientific studies show that there are many known nanoformulations with curcumin. They can be found in nanosuspensions, nanoparticles, nanoemulsions, solid lipid particles, nanocapsules, nanospheres, and liposomes. These formulations can improve CUR bioavailability and can effectively be used as adjuvants in several inflammatory and immune-mediated diseases such as atheroma plaque formation, RA, dementia, AD, PD, MS, IBD, psoriasis, epilepsy, COVID-19, and can be used as potent anti-fibrotic adjuvants in fibrotic liver disease.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Gabriel Magno de Carvalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Bárbara de Oliveira Zanuso
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Maria Eduardo Figueira
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Daiene Santos Buglio
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília, São Paulo 17500-000, Brazil
- Correspondence: ; Tel.: +55-14-99655-3190
| |
Collapse
|
19
|
Zhang X, Zhao X, Hua Z, Xing S, Li J, Fei S, Tan M. ROS-triggered self-disintegrating and pH-responsive astaxanthin nanoparticles for regulating the intestinal barrier and colitis. Biomaterials 2023; 292:121937. [PMID: 36495803 DOI: 10.1016/j.biomaterials.2022.121937] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
Smart delivery systems with stimuli-responsive capability are able to improve the bioaccessibility through increasing the solubility, physicochemical stability and biocompatibility of bioactive compounds. In this study, the astaxanthin nanoparticles with reactive oxygen species (ROS) and pH dual-response function were design and constructed using poly (propylene sulfide) covalently modified sodium alginate as carriers based on ultrasonic assisted self-assembly strategy. Atomic force microscope and scanning electron microscope analysis showed that the nanoparticles were spherical in shape with a size of around 260 nm. Meanwhile, the astaxanthin nanoparticles showed both pH and ROS stimuli-responsive release characteristics. In vitro cell experiments showed that astaxanthin nanoparticles significantly inhibited the production of ROS and mitochondrial depolarization induced by oxidative stress. In vivo colitis experiment of mice revealed that astaxanthin nanoparticles could significantly relieve colitis, protect the integrity of colon tissue and restore the expression of tight junction proteins ZO-1 and occludin. The abundance of Lactobacillus and Lachnospiraceae, and the ratio of Firmicutes/Bacteroidota of gut microbiota were significantly improved after intervention of the stimuli-responsive astaxanthin nanoparticles. This work provided a simple strategy for constructing ROS/pH dual response delivery system, which provided an experimental basis for improving the oral bioavailability of hydrophobic active compounds.
Collapse
Affiliation(s)
- Xuedi Zhang
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Xue Zhao
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Zheng Hua
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Shanghua Xing
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Jiaxuan Li
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Siyuan Fei
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China.
| |
Collapse
|
20
|
Zhong Y, Xiao Q, Kang Z, Huang J, Ge W, Wan Q, Wang H, Zhou W, Zhao H, Liu D. Astragalus polysaccharide alleviates ulcerative colitis by regulating the balance of Tfh/Treg cells. Int Immunopharmacol 2022; 111:109108. [PMID: 35926271 DOI: 10.1016/j.intimp.2022.109108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 11/23/2022]
Abstract
The immunomodulatory function of natural active ingredients has long been a focus of scientific research, with recent hotspots reporting targeted modulation of the follicular helper T cells (Tfh)/regulatory T cells (Treg) balance as an emerging strategy for the treatment of ulcerative colitis (UC). Here, dextran sodium sulfate induced mice UC and Astragalus polysaccharide (APS, 200 mg/kg/day) was administered simultaneously. In this study, APS effectively alleviated colitis in mice by improving survival rate, disease activity index (DAI), the change rate of body weight, colonic length and weight, and histopathological injury of the colon. Moreover, APS regulated the expression of inflammatory cytokines interleukin (IL)-2, IL-6, IL-12p70, IL-23, Tumour necrosis factor (TNF)-ɑ, and transforming growth factor (TGF)-β1 in colonic tissues of colitis mice. Importantly, APS significantly downregulated Tfh cell and the expression of its related nuclear transcription factors Blimp-1 and Bcl-6, and cytokine IL-21. Meanwhile, APS regulated the differentiation of Tfh subpopulations in colitis mice, with Tfh10 and Tfr significantly upregulated while Tfh1, Tfh17, and Tfh21 significantly downregulated. In addition, APS significantly upregulated Treg cells and the levels of its associated nuclear transcription factor Foxp3, and cytokine IL-10 in colitis mice. In conclusion, APS effectively alleviated UC by reshaping the balance of Tfh/Treg cells.
Collapse
Affiliation(s)
- Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Qiuping Xiao
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330004, Jiangxi Province, China
| | - Zengping Kang
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wei Ge
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Qi Wan
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Haiyan Wang
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wen Zhou
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Nanchang Medical College, Nanchang 330004, Jiangxi Province, China
| | - Haimei Zhao
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|
21
|
Zhong Y, Liu W, Xiong Y, Li Y, Wan Q, Zhou W, Zhao H, Xiao Q, Liu D. Astragaloside Ⅳ alleviates ulcerative colitis by regulating the balance of Th17/Treg cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154287. [PMID: 35752072 DOI: 10.1016/j.phymed.2022.154287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/01/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Restoring immune homeostasis by targeting the Th17/Treg response is a potentially valuable therapeutic strategy for ulcerative colitis (UC). Astragaloside IV (AS-Ⅳ) is a phytochemical naturally occurring in Astragalus membranaceus that has good anti-inflammatory, anti-oxidant and anti-stress properties. However, the effects of AS-IV on the homeostasis of Th17/Treg cells in colitis mice remains unknown. PURPOSE To investigate the protective effects and potential immunomodulatory mechanisms of AS-IV on UC. METHODS This study was constructed for DSS-induced acute colitis and recurrent colitis, with AS-IV administered prophylactically and therapeutically, respectively. The balance of Th17/Treg cells was analyzed by flow cytometry, their specific nuclear transcription factors were detected by RT-PCR as well as their secreted inflammatory cytokines were detected by ELISA and RT-PCR. Notch signaling-related proteins were detected by RT-PCR and Western blotting. Oxidative stress indicators were measured by biochemical technology. RESULTS In this study, AS-IV treatment not only effectively prevented and alleviated the clinical symptoms of DSS-induced colitis mice, including weight loss, DAI soaring, colon length shortening and colon weight gain, but also significantly improved ulcer formation, inflammatory cell infiltration and index, and regulated the expression of inflammatory cytokines in colon tissues. Importantly, the efficacy of high-dose AS-IV (100 mg/kg/day) in mice with recurrent colitis in this study was comparable to that of 5-ASA. AS-IV early administration was able to reshape the homeostasis of Th17/Treg cells in mice with acute colitis; meanwhile, AS-IV inhibited Th17 cell responses and promoted Treg cell responses in mice with recurrent colitis. Moreover, AS-IV not only inhibited the activation of Notch signaling pathway in colitis mice, but also prevented and ameliorated DSS-induced oxidative stress injury. CONCLUSION In conclusion, AS-IV effectively prevented and alleviated UC by reshaping Th17/Treg cell homeostasis and anti-oxidative stress.
Collapse
Affiliation(s)
- Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Jiangxi 330004, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Wenjun Liu
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi 330004, China
| | - Yanxia Xiong
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi 330004, China
| | - Yingmeng Li
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi 330004, China
| | - Qi Wan
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Wen Zhou
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China; Nanchang Medical college, Nanchang, Jiangxi 330004, China
| | - Haimei Zhao
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Jiangxi 330004, China
| | - Qiuping Xiao
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi 330004, China.
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Jiangxi 330004, China.
| |
Collapse
|
22
|
Changes of Intestinal Flora and Its Relationship with Nutritional Status for Patients with Cancer Pain. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5188202. [PMID: 36035282 PMCID: PMC9402348 DOI: 10.1155/2022/5188202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022]
Abstract
Objective. To study the changes in the intestinal flora and its relationship with nutritional status for patients with cancer pain. Methods. A prospective research method was adopted. One hundred twenty cancer patients with cancer pain were selected as the research objects, who were treated in our hospital from June 2019 to June 2020, and 120 cancer patients without cancer pain were selected as the control group, who were treated in the same period. The differences of the intestinal flora and nutritional status of patients with different severity between the observation group and the control group were compared to analyze the changes of intestinal flora in patients with cancer pain and its correlation with nutritional status. Results. Hemoglobin (HB) (
,
), albumin (ALB) (
,
), prealbumin (PAB) (
,
), and total protein (TP) (
,
) in the observation group were significantly lower than those in the control group. There were statistically significant differences in HB (
,
), ALB (
,
), PAB (
,
), and TP (
,
) among patients with cancer pain of different severity. Through these two comparisons, their nutritional indicators showed a significant downward trend with the increase in the severity for cancer pain patients; the levels of Lactobacillus (
,
), Bifidobacterium (
,
), Enterococcus (
,
), and Eubacterium (
,
) in the observation group were significantly lower than those in the control group. There were statistically significant differences in the levels of Lactobacillus (
,
), Bifidobacterium (
,
), Enterococcus (
,
), and Eubacterium (
,
) among patients with cancer pain of different severity. After pairwise comparison, their beneficial intestinal bacteria were significantly lower than those in the control group with the increase in pain in cancer pain patients. Nitric oxide (NO) (
,
), galectin-3 (
,
), occludin (OCLN) (
,
), galectin-1 (
,
), zonula occludens protein 1 (ZO-1) (
,
), and cingulin (
,
) in the observation group were significantly lower than those in the control group. There were statistically significant differences in NO, galectin-3, OCLN, galectin-1, ZO-1, and cingulin for patients with cancer pain of different severity. By comparison, the NO, galectin-3, OCLN, galectin-1, ZO-1, and cingulin of the patients showed a significant downward trend with the aggravation of cancer pain symptoms. Through correlation analysis, the nutritional indicators of patients were positively correlated with intestinal microorganisms and intestinal barrier function. Conclusion. There was a significant correlation between the changes in intestinal flora and nutritional status for patients with cancer pain, which could be used as an important basis for improving the treatment of cancer pain.
Collapse
|
23
|
Zhou Z, Yu S, Cui L, Shao K, Pang H, Wang Z, He N, Li S. Isomaltulose alleviates acute colitis via modulating gut microbiota and the Treg/Th17 balance in mice. Food Funct 2022; 13:8572-8584. [PMID: 35894244 DOI: 10.1039/d2fo01157c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Food-grade isomaltulose exhibits significant modulation of gut microbiota and its metabolites in healthy populations. This study further explored the preventive therapeutic effect and anti-colitis potential of isomaltulose on dextran sulfate sodium-induced colitis in mice. Our results suggested that isomaltulose played a significant role in preventing colon shortening, reducing intestinal epithelial destruction and inhibiting inflammatory cell infiltration. Meanwhile, the isomaltulose supplement greatly reduced the production of pro-inflammatory cytokines and restored the balance between T helper type 17 (Th17) cells and regulatory T (Treg) cells. Pathway enrichment analysis for differentially expressed genes (DEGs) also indicated that the anti-inflammatory effect of isomaltulose was closely related to intestinal immunity. Moreover, the disturbed gut microbiota in ulcerative colitis (UC) was partially restored after treatment with isomaltulose. These results suggest that isomaltulose is a promising therapeutic agent for the prevention and adjunctive treatment of UC by maintaining intestinal immune homeostasis and remodeling the gut microbiota.
Collapse
Affiliation(s)
- Zihan Zhou
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Shengnan Yu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Luwen Cui
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Kaidi Shao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Hao Pang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Zhipeng Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
24
|
Chen S, Zhu H, Luo Y. Chitosan-based oral colon-specific delivery systems for polyphenols: recent advances and emerging trends. J Mater Chem B 2022; 10:7328-7348. [PMID: 35766297 DOI: 10.1039/d2tb00874b] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oral colon-targeted delivery systems (OCDSs) have attracted great attention in the delivery of active compounds targeted to the colon for the treatment of colon and non-colon diseases with the advantages of enhanced efficacy and reduced side effects. Chitosan, the second-most abundant biopolymer next to cellulose, has great biocompatibility, is non-toxic, is sensitive to colonic flora and shows strong adhesion to colonic mucus, making it an ideal biomaterial candidate for the construction of OCDSs. Being rich in functional groups, the chitosan structure is easily modified, both physically and chemically, for the fabrication of delivery systems with diverse geometries, including nanoparticles, microspheres/microparticles, and hydrogels, that are resistant to the harsh environment of the upper gastrointestinal tract (GIT). This review offers a detailed overview of the preparation of chitosan-based delivery systems as the basis for building OCDSs. A variety of natural polyphenols with potent biological activities are used to treat diseases of the colon, or to be metabolized as active ingredients by colonic microorganisms to intervene in remote organ diseases after absorption into the circulation. However, the poor solubility of polyphenols limits their application, and the acidic environment of the upper GIT and various enzymes in the small intestine disrupt their structure and activity. As a result, the development of OCDSs for polyphenols has become an emerging and popular area of current research in the past decade. Thus, the second objective of this review is to systematically summarize the most recent research findings in this area and shed light on the future development of chitosan-based OCDSs for nutritional and biomedical applications.
Collapse
Affiliation(s)
- Sunni Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Honglin Zhu
- Nanotechnology and Biodelivery Laboratory, Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | - Yangchao Luo
- Nanotechnology and Biodelivery Laboratory, Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
25
|
Liu N, Wang H, Yang Z, Zhao K, Li S, He N. The role of functional oligosaccharides as prebiotics in ulcerative colitis. Food Funct 2022; 13:6875-6893. [PMID: 35703137 DOI: 10.1039/d2fo00546h] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The incidence rate of ulcerative colitis (UC) has increased significantly over the past decades and it places an increasing burden on health and social systems. The current studies on UC implicate a strong correlation between host gut microbiota immunity and the pathogenesis of UC. Meanwhile, more and more functional oligosaccharides have been reported as prebiotics to alleviate UC, since many of them can be metabolized by gut microbiota to produce short-chain fatty acids (SCFAs). The present review is focused on the structure, sources and specific applications of various functional oligosaccharides related to the prevention and treatment of UC. The available evidence for the usage of functional oligosaccharides in UC treatment are summarized, including fructo-oligosaccharides (FOS), galacto-oligosaccharides (GOS), chito-oligosaccharides (COS), alginate-oligosaccharides (AOS), xylooligosaccharides (XOS), stachyose and inulin.
Collapse
Affiliation(s)
- Nian Liu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Haoyu Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Kunyi Zhao
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
26
|
Rahiman N, Markina YV, Kesharwani P, Johnston TP, Sahebkar A. Curcumin-based nanotechnology approaches and therapeutics in restoration of autoimmune diseases. J Control Release 2022; 348:264-286. [PMID: 35649486 DOI: 10.1016/j.jconrel.2022.05.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases usually arise as a result of an aberrant immune system attack on normal tissues of the body, which leads to a cascade of inflammatory reactions. The immune system employs different types of protective and anti-inflammatory cells for the regulation of this process. Curcumin is a known natural anti-inflammatory agent that inhibits pathological autoimmune processes by regulating inflammatory cytokines and their associated signaling pathways in immune cells. Due to the unstable nature of curcumin and its susceptibility to either degradation, or metabolism into other chemical entities (i.e., metabolites), encapsulation of this agent into various nanocarriers would appear to be an appropriate strategy for attaining greater beneficial effects from curcumin as it pertains to immunomodulation. Many studies have focused on the design and development of curcumin nanodelivery systems (micelles, dendrimers, and diverse nanocarriers) and are summarized in this review in order to obtain greater insight into novel drug delivery systems for curcumin and their suitability for the management of autoimmune diseases.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad, Iran
| | - Yuliya V Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBI "Petrovsky National Research Center of Surgery", 3 Tsyurupy Str., 117418, Moscow, Russia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran..
| |
Collapse
|
27
|
Zhong Y, Xiao Q, Li S, Chen L, Long J, Fang W, Yu F, Huang J, Zhao H, Liu D. Bupi Yichang Pill alleviates dextran sulfate sodium-induced ulcerative colitis in mice by regulating the homeostasis of follicular helper T cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154091. [PMID: 35395566 DOI: 10.1016/j.phymed.2022.154091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/07/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Follicular helper T (Tfh) cells-based therapy represents a new treatment option for inflammatory bowel disease. Bupi Yichang Pill (BPYCP), a traditional Chinese formula for the treatment of dysentery and diarrhea, exhibits potential anti-inflammatory activities in treating various kinds of inflammation. However, its anti-inflammatory effect on colitis and the underlying mechanisms remain unknown. PURPOSE To explore the protective role and underlying immunomodulatory effects of BPYCP in the treatment of UC. METHODS The dextran sodium sulfate (DSS) free-drinking method induced UC in C57BL/6 mice, and BPYCP was orally administrated at a dosage of 1.5, 3.0, or 6.0 g/kg/day. Throughout the experimental period, the effects of BPYCP on DSS-induced clinical symptoms and disease activity index (DAI) were monitored and analyzed. Hematoxylin-eosin staining was used to observe the histopathological injury of the colon. Flow cytometry was used to detect the levels of Tfh cells, Tfh cell subpopulations, and memory Tfh cells. ELISA, Western blot, and qRT-PCR were used to detect the expression of inflammatory cytokines and Tfh cell-related biomarkers. RESULTS Medium and high dosages of BPYCP effectively alleviated DSS-induced experimental colitis with increased body weight, survival rate and colonic length, and decreased DAI, colonic weight, and colonic index, as well as less ulcer formation and inflammatory cell infiltration, increased anti-inflammatory cytokine IL-10, and decreased pro-inflammatory cytokines IL-17A, IL-6, and TNF-ɑ. Moreover, BPYCP administration significantly decreased the percentage of Tfh cells and the expression of Tfh markers ICOS, PD-1 and Bcl-6 in the mesenteric lymph nodes of colitis mice. In addition, BPYCP treatment obviously decreased the percentages of Tfh1, Tfh17 and Tem-Tfh cells and upregulated Tfr cells in colitis mice. However, there were no significant regulatory effects of BPYCP on Tfh cell response in normal mice. CONCLUSION Taken together, these results demonstrated a protective effect of BPYCP against DSS-induced experimental colitis by regulating Tfh cell homeostasis.
Collapse
Affiliation(s)
- Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qiuping Xiao
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, 330004, Jiangxi Province, China
| | - Shanshan Li
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Liling Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jian Long
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Weiyan Fang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Feihao Yu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Jiaqi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Haimei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| |
Collapse
|
28
|
Liu R, Luo C, Pang Z, Zhang J, Ruan S, Wu M, Wang L, Sun T, Li N, Han L, Shi J, Huang Y, Guo W, Peng S, Zhou W, Gao H. Advances of nanoparticles as drug delivery systems for disease diagnosis and treatment. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Li S, Zhang H, Chen K, Jin M, Vu SH, Jung S, He N, Zheng Z, Lee MS. Application of chitosan/alginate nanoparticle in oral drug delivery systems: prospects and challenges. Drug Deliv 2022; 29:1142-1149. [PMID: 35384787 PMCID: PMC9004504 DOI: 10.1080/10717544.2022.2058646] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oral drug delivery systems (ODDSs) have various advantages of simple operation and few side effects. ODDSs are highly desirable for colon-targeted therapy (e.g. ulcerative colitis and colorectal cancer), as they improve therapeutic efficiency and reduce systemic toxicity. Chitosan/alginate nanoparticles (CANPs) show strong electrostatic interaction between the carboxyl group of alginates and the amino group of chitosan which leads to shrinkage and gel formation at low pH, thereby protecting the drugs from the gastrointestinal tract (GIT) and aggressive gastric environment. Meanwhile, CANPs as biocompatible polymer, show intestinal mucosal adhesion, which could extend the retention time of drugs on inflammatory sites. Recently, CANPs have attracted increasing interest as colon-targeted oral drug delivery system for intestinal diseases. The purpose of this review is to summarize the application and treatment of CANPs in intestinal diseases and insulin delivery. And then provide a future perspective of the potential and development direction of CANPs as colon-targeted ODDSs.
Collapse
Affiliation(s)
- Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China.,Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul, Korea
| | - Hui Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Kaiwei Chen
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Mengfei Jin
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Son Hai Vu
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul, Korea.,Institute of Applied Sciences, Ho Chi Minh City University of Technology HUTECH, Ho Chi Minh City, Viet Nam
| | - Samil Jung
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul, Korea
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhou Zheng
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resource, Qingdao, China
| | - Myeong-Sok Lee
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|
30
|
Li X, Yang Y, Wang Z, Ju H, Fu X, Zou L, Li M, Xue Q, Ma H, Meng Y, Zhao L, Qi H, Yu T. Multistage-Responsive Nanocomplexes Attenuate Ulcerative Colitis by Improving the Accumulation and Distribution of Oral Nucleic Acid Drugs in the Colon. ACS APPLIED MATERIALS & INTERFACES 2022; 14:2058-2070. [PMID: 34978415 DOI: 10.1021/acsami.1c21595] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Oral gene therapy has emerged as a potential optimal treatment for ulcerative colitis (UC). Nucleic acid drugs possessing versatility can not only inhibit inflammation but realize colon mucosal healing, fulfilling the clinical objective of UC therapy. However, the effective accumulation and distribution of oral nucleic acid drugs in the colon remain a considerable challenge. Furthermore, current delivery systems pay more attention to the accumulation of nucleic acid drugs in the colon, while the distribution of nucleic acid drugs in the colon, which plays a key role in the UC treatment, never catches the attention of researchers. Here, we used miR-320 as a model nucleic acid drug to develop a kind of multistage-responsive nanocomplexes (MSNs) based on polymeric nanocapsules and alginate. MSNs possess the pH responsiveness in the stomach, the enzyme responsiveness in the colonic lumen, and the redox responsiveness in the cytoplasm. In vivo imaging results showed that MSNs reach the colon within 2 h and effectively release miR-320 nanocapsules in the colonic lumen. The nanocapsules can further deliver miR-320 to the submucosal layer and even the muscular layer. Moreover, MSNs decreased the activity of myeloperoxidase and proinflammatory cytokines and exhibited anti-inflammatory activity by inhibiting the phosphorylation of IκBα and AKT, reducing colonic inflammation and enhancing mucosal repair. Therefore, MSNs can successfully alleviate UC by improving the accumulation and distribution of oral nucleic acid drugs in the colon, promoting the clinical translational application of nucleic acid drugs in the treatment of UC.
Collapse
Affiliation(s)
- Xiaoxin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, Qingdao 266021, China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hui Ju
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lu Zou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Qianqian Xue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Huibo Ma
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yuanyuan Meng
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Liang Zhao
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
31
|
Wang H, Liu N, Yang Z, Zhao K, Pang H, Shao K, Zhou Z, Li S, He N. Preventive Effect of Pectic Oligosaccharides on Acute Colitis Model Mice: Modulating Epithelial Barrier, Gut Microbiota and Treg/Th17 Balance. Food Funct 2022; 13:9999-10012. [DOI: 10.1039/d2fo01448c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pectin as dietary fiber supplements has shown emerging potential in clinical ulcerative colitis (UC) adjuvant therapy. In this study, the preventive and prebiotic effect of enzymatic-degraded pectic oligosaccharides (POS) was...
Collapse
|