1
|
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] [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
|
2
|
Parveen S, Alqahtani AS, Aljabri MY, Bajonaid A, Khan SS, Hassan AAHAA, Dawood T. Nationwide exploration: assessing oral microbiome knowledge among dental professionals in Saudi Arabia and its implications for oral health care. BMC Oral Health 2024; 24:1028. [PMID: 39217310 PMCID: PMC11366131 DOI: 10.1186/s12903-024-04770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The relationship between the microbiome and oral health is intricate, yet there is a lack of comprehensive knowledge regarding the microbiome's impact on oral health. Integrating knowledge regarding the oral microbiome and its significance in oral and systemic diseases holds profound implications for dental professionals in patient care and professional development. This study assessed dental professionals' oral microbiome comprehension and knowledge levels in Saudi Arabia and its implications for oral healthcare. METHODS Data were gathered using a cross-sectional design by administering a comprehensive online questionnaire to 253 dental professionals from diverse demographic backgrounds. The questionnaire, administered in English, was divided into four sections: (1) Microbiome awareness and understanding, (2) Diet, nutrition, and microbiome relationship, (3) Microbiome and oral and systemic diseases, and (4) Counselling, education, and implications. Statistical analyses were used to identify and understand underlying patterns, including descriptive statistics, chi-squared tests, ANOVA, and post hoc tests. The Spearman rank correlation coefficient was applied to assess self-rated knowledge. RESULTS Of the 253 participants, 94.6% were familiar with the term "microbiome." Merely 13% of participants considered the oral microbiome to be the second most diverse, following the gut microbiome. About 39.9% of participants knew the connection between oral mucosal diseases and the oral microbiome. Furthermore, only 6.7% thought there was a connection between systemic diseases and the oral microbiome. Participant comprehension of oral microbiome questions averaged 9.19 out of 13, with 83.7% scoring "good". There were significant differences in knowledge scores among dental specializations (F = 7.082, P < 0.001) and years of professional experience (F = 4.755, P = 0.003). Significantly, 53.8% of participants had uncertain self-perceptions of their knowledge of the oral microbiome, while only 0.8% rated their understanding as 'very good'. CONCLUSION Our findings reveal that dental professionals have varying levels of awareness and comprehension of the oral microbiome. Despite widespread awareness, understanding its diversity and implications for oral and systemic health remains limited. It is essential to address these gaps in knowledge through future research and educational interventions, considering the vital part that dental professionals play in promoting oral health through personalised dietary recommendations, lifestyle changes, and hygiene practices. These initiatives may promote a robust oral microbial community, enhance patient outcomes, and advance oral healthcare locally and globally.
Collapse
Affiliation(s)
- Sameena Parveen
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | - Ahmed Shaher Alqahtani
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Mohammed Y Aljabri
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Amal Bajonaid
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Samar Saeed Khan
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | | | - Tazeen Dawood
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
3
|
Pärnänen P, Räisänen IT, Sorsa T. Oral Anti-Inflammatory and Symbiotic Effects of Fermented Lingonberry Juice-Potential Benefits in IBD. Nutrients 2024; 16:2896. [PMID: 39275212 PMCID: PMC11397234 DOI: 10.3390/nu16172896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Microbial dysbiosis may manifest as inflammation both orally and in the gastrointestinal tract. Altered oral and gut microbiota composition and decreased diversity have been shown in inflammatory bowel disease (IBD) and periodontal disease (PD). Recent studies have verified transmission of oral opportunistic microbes to the gut. Prebiotics, probiotics, or dietary interventions are suggested to alleviate IBD symptoms in addition to medicinal treatment. Lingonberries contain multiple bioactive molecules, phenolics, which have a broad spectrum of effects, including antimicrobial, anti-inflammatory, antioxidant, anti-proteolytic, and anti-cancer properties. An all-natural product, fermented lingonberry juice (FLJ), is discussed as a potential natural anti-inflammatory substance. FLJ has been shown in clinical human trials to promote the growth of oral lactobacilli, and inhibit growth of the opportunistic oral pathogens Candida, Streptococcus mutans, and periodontopathogens, and decrease inflammation, oral destructive proteolysis (aMMP-8), and dental microbial plaque load. Lactobacilli are probiotic and considered also beneficial for gut health. Considering the positive outcome of these oral studies and the fact that FLJ may be swallowed safely, it might be beneficial also for the gut mucosa by balancing the microbiota and reducing proteolytic inflammation.
Collapse
Affiliation(s)
- Pirjo Pärnänen
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Ismo T Räisänen
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
4
|
Ge J, Li M, Yao J, Guo J, Li X, Li G, Han X, Li Z, Liu M, Zhao J. The potential of EGCG in modulating the oral-gut axis microbiota for treating inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155643. [PMID: 38820660 DOI: 10.1016/j.phymed.2024.155643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 06/02/2024]
Abstract
Inflammatory bowel disease (IBD) is a recurrent chronic intestinal disorder that includes ulcerative colitis (UC) and Crohn's disease (CD). Its pathogenesis involves intricate interactions between pathogenic microorganisms, native intestinal microorganisms, and the intestinal immune system via the oral-gut axis. The strong correlation observed between oral diseases and IBD indicates the potential involvement of oral pathogenic microorganisms in IBD development. Consequently, therapeutic strategies targeting the proliferation, translocation, intestinal colonization and exacerbated intestinal inflammation of oral microorganisms within the oral-gut axis may partially alleviate IBD. Tea consumption has been identified as a contributing factor in reducing IBD, with epigallocatechin gallate (EGCG) being the primary bioactive compound used for IBD treatment. However, the precise mechanism by which EGCG mediates microbial crosstalk within the oral-gut axis remains unclear. In this review, we provide a comprehensive overview of the diverse oral microorganisms implicated in the pathogenesis of IBD and elucidate their colonization pathways and mechanisms. Subsequently, we investigated the antibacterial properties of EGCG and its potential to attenuate microbial translocation and colonization in the gut, emphasizing its role in attenuating exacerbations of IBD. We also elucidated the toxic and side effects of EGCG. Finally, we discuss current strategies for enhancing EGCG bioavailability and propose novel multi-targeted nano-delivery systems for the more efficacious management of IBD. This review elucidates the role and feasibility of EGCG-mediated modulation of the oral-gut axis microbiota in the management of IBD, contributing to a better understanding of the mechanism of action of EGCG in the treatment of IBD and the development of prospective treatment strategies.
Collapse
Affiliation(s)
- Jiaming Ge
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengyuan Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingwen Yao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinling Guo
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiankuan Li
- Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Gang Li
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Xiangli Han
- Department of Geriatric, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin 300450, China
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ming Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China.
| | - Jing Zhao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
5
|
Pärnänen P, Suojanen J, Laine M, Sorsa T, Ranki A. Long-term remission of candidiasis with fermented lingonberry mouth rinse in an adult patient with APECED. Int J Infect Dis 2024; 144:107066. [PMID: 38649005 DOI: 10.1016/j.ijid.2024.107066] [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: 02/12/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
We report a long-term remission in candidiasis in a 57-year-old Finnish female with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) suffering from recurrent oral, esophageal, gastric, vaginal, and anal candidiasis since childhood. Candidiasis treatment with antifungal medicines fluconazole, itraconazole, posaconazole, voriconazole, caspofungin, nystatin, or amphotericin-B during 2008-2021 had variable effects and intermittent development of antifungal resistance and hospital periods. The patient started using fermented lingonberry juice (FLJ) as a mouth rinse daily in April 2021. No symptoms or mucosal signs of candidiasis in any part of the digestive system or vaginal area have been noticed during this exceptionally long-term 2 ½ year remission in candidiasis without antifungal medications.
Collapse
Affiliation(s)
- Pirjo Pärnänen
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Juho Suojanen
- Helsinki University Hospital, Cleft Palate and Craniofacial Centre, Helsinki, Finland; Department of Maxillofacial Surgery, Päijät-Häme Central Hospital, Joint Authority for Health and Wellbeing, Lahti, Finland; Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Laine
- Porvoo Hospital, Department of Gastrointestinal Surgery, Hospital District of Helsinki and Uusimaa, Porvoo, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Annamari Ranki
- Department of Dermatology and Allergology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
6
|
Chu X, Chen X, Zhang H, Wang Y, Guo H, Chen Y, Liu X, Zhu Z, He Y, Ding X, Wang Q, Zheng C, Cao X, Yang H, Qian J. Association of diet and outdoor time with inflammatory bowel disease: a multicenter case-control study using propensity matching analysis in China. Front Public Health 2024; 12:1368401. [PMID: 38952728 PMCID: PMC11215971 DOI: 10.3389/fpubh.2024.1368401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/09/2024] [Indexed: 07/03/2024] Open
Abstract
Objective To investigate the association between dietary and some other environmental factors and the risk of inflammatory bowel diseases (IBD) in Chinese population. Materials and methods A multicenter case-control study was conducted involving 11 hospitals across China. A total of 1,230 subjects were enrolled consecutively, and diet and environmental factor questionnaires were collected. IBD patients were matched with healthy controls (HC) using propensity-score matching (PSM) at a 1:1 ratio with a caliper value of 0.02. Multivariate conditional logistic regression analyses were performed to evaluate the associations between diet, environmental factors, and IBD. Results Moderate alcohol and milk consumption, as well as daily intake of fresh fruit, were protective factors for both Crohn's disease (CD) and ulcerative colitis (UC). Conversely, the consumption of eggs and chocolate increased the risk of IBD. Outdoor time for more than 25% of the day was a protective factor only for CD. In eastern regions of China, CD patients had higher egg consumption and less outdoor time, while UC patients consumed more chocolate. IBD patients from urban areas or with higher per capita monthly income consumed more fruit, eggs, and chocolate. Conclusions This study reveals an association between specific foods, outdoor time, and the emergence of IBD in the Chinese population. The findings emphasize the importance of a balanced diet, sufficient outdoor time and activities, and tailored prevention strategies considering regional variations.
Collapse
Affiliation(s)
- Xiaotian Chu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuanfu Chen
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huimin Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yufang Wang
- Department of Gastroenterology, Sichuan University West China Hospital, Chengdu, China
| | - Hong Guo
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yan Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yao He
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xueli Ding
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qunying Wang
- Gastroenterology and Hepatology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Changqing Zheng
- Department of Gastroenterology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Chaudhary PP, Kaur M, Myles IA. Does "all disease begin in the gut"? The gut-organ cross talk in the microbiome. Appl Microbiol Biotechnol 2024; 108:339. [PMID: 38771520 PMCID: PMC11108886 DOI: 10.1007/s00253-024-13180-9] [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: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
The human microbiome, a diverse ecosystem of microorganisms within the body, plays pivotal roles in health and disease. This review explores site-specific microbiomes, their role in maintaining health, and strategies for their upkeep, focusing on oral, lung, vaginal, skin, and gut microbiota, and their systemic connections. Understanding the intricate relationships between these microbial communities is crucial for unraveling mechanisms underlying human health. Recent research highlights bidirectional communication between the gut and distant microbiome sites, influencing immune function, metabolism, and disease susceptibility. Alterations in one microbiome can impact others, emphasizing their interconnectedness and collective influence on human physiology. The therapeutic potential of gut microbiota in modulating distant microbiomes offers promising avenues for interventions targeting various disorders. Through interdisciplinary collaboration and technological advancements, we can harness the power of the microbiome to revolutionize healthcare, emphasizing microbiome-centric approaches to promote holistic well-being while identifying areas for future research.
Collapse
Affiliation(s)
- Prem Prashant Chaudhary
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Mahaldeep Kaur
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
8
|
Sidiropoulos T, Dovrolis N, Katifelis H, Michalopoulos NV, Kokoropoulos P, Arkadopoulos N, Gazouli M. Dysbiosis Signature of Fecal Microbiota in Patients with Pancreatic Adenocarcinoma and Pancreatic Intraductal Papillary Mucinous Neoplasms. Biomedicines 2024; 12:1040. [PMID: 38791002 PMCID: PMC11117863 DOI: 10.3390/biomedicines12051040] [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: 04/07/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic cancer (PC) ranks as the seventh leading cause of cancer-related deaths, with approximately 500,000 new cases reported in 2020. Existing strategies for early PC detection primarily target individuals at high risk of developing the disease. Nevertheless, there is a pressing need to identify innovative clinical approaches and personalized treatments for effective PC management. This study aimed to explore the dysbiosis signature of the fecal microbiota in PC and potential distinctions between its Intraductal papillary mucinous neoplasm (IPMN) and pancreatic ductal adenocarcinoma (PDAC) phenotypes, which could carry diagnostic significance. The study enrolled 33 participants, including 22 diagnosed with PDAC, 11 with IPMN, and 24 healthy controls. Fecal samples were collected and subjected to microbial diversity analysis across various taxonomic levels. The findings revealed elevated abundances of Firmicutes and Proteobacteria in PC patients, whereas healthy controls exhibited higher proportions of Bacteroidota. Both LEfSe and Random Forest analyses indicated the microbiome's potential to effectively distinguish between PC and healthy control samples but fell short of differentiating between IPMN and PDAC samples. These results contribute to the current understanding of this challenging cancer type and highlight the applications of microbiome research. In essence, the study provides clear evidence of the gut microbiome's capability to serve as a biomarker for PC detection, emphasizing the steps required for further differentiation among its diverse phenotypes.
Collapse
Affiliation(s)
- Theodoros Sidiropoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Nikolas Dovrolis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| | - Hector Katifelis
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| | - Nikolaos V. Michalopoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Panagiotis Kokoropoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Nikolaos Arkadopoulos
- 4th Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (T.S.); (N.V.M.); (P.K.); (N.A.)
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.D.); (H.K.)
| |
Collapse
|
9
|
Fang X, Feng J, Zhu X, Feng D, Zheng L. Plant-derived vesicle-like nanoparticles: A new tool for inflammatory bowel disease and colitis-associated cancer treatment. Mol Ther 2024; 32:890-909. [PMID: 38369751 PMCID: PMC11163223 DOI: 10.1016/j.ymthe.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/03/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
Long-term use of conventional drugs to treat inflammatory bowel diseases (IBD) and colitis-associated cancer (CAC) has an adverse impact on the human immune system and easily leads to drug resistance, highlighting the urgent need to develop novel biotherapeutic tools with improved activity and limited side effects. Numerous products derived from plant sources have been shown to exert antibacterial, anti-inflammatory and antioxidative stress effects. Plant-derived vesicle-like nanoparticles (PDVLNs) are natural nanocarriers containing lipids, protein, DNA and microRNA (miRNA) with the ability to enter mammalian cells and regulate cellular activity. PDVLNs have significant potential in immunomodulation of macrophages, along with regulation of intestinal microorganisms and friendly antioxidant activity, as well as overcoming drug resistance. PDVLNs have utility as effective drug carriers and potential modification, with improved drug stability. Since immune function, intestinal microorganisms, and antioxidative stress are commonly targeted key phenomena in the treatment of IBD and CAC, PDVLNs offer a novel therapeutic tool. This review provides a summary of the latest advances in research on the sources and extraction methods, applications and mechanisms in IBD and CAC therapy, overcoming drug resistance, safety, stability, and clinical application of PDVLNs. Furthermore, the challenges and prospects of PDVLN-based treatment of IBD and CAC are systematically discussed.
Collapse
Affiliation(s)
- Xuechun Fang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junjie Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xingcheng Zhu
- Medical Laboratory Department, Second People's Hospital, Qujing 655000, China
| | - Dan Feng
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510182, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
10
|
Jiang N, Liu Z, Wang H, Zhang L, Li M, Li G, Li C, Wang B, Zhao C, Liu L. Alterations in metabolome and microbiome: new clues on cathelicidin-related antimicrobial peptide alleviates acute ulcerative colitis. Front Microbiol 2024; 15:1306068. [PMID: 38380090 PMCID: PMC10877057 DOI: 10.3389/fmicb.2024.1306068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/26/2024] [Indexed: 02/22/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic and recurrent inflammatory disease of the gastrointestinal tract. This study aimed to determine the effect of cathelicidin-related antimicrobial peptide (Cramp) on dextran sulfate sodium (DSS)-induced acute experimental colitis in mice and to investigate the underlying mechanisms. Acute UC was induced in C57BL/6 mice with 3% DSS for 7 days, 4 mg/kg b.w. synthetic Cramp peptide was administrated once daily starting on day 4 of the experimental period. Mice were evaluated for body weight, colon length, colon histopathology, and inflammatory cytokines in colon tissue. Using 16 s rRNA sequencing, the composition structure of gut microbiota was characterized. Metabolomic profiling of the serum was performed. The results showed that DSS treatment significantly induced intestinal damage as reflected by disease activity index, histopathological features, and colon length, while Cramp treatment significantly prevented these trends. Meanwhile, Cramp treatment decreased the levels of inflammatory cytokines in both serum and colonic tissue on DSS-induced colitis. It was also observed that DSS damaged the integrity of the intestinal epithelial barrier, whereas Cramp also played a protective role by attenuating these deteriorated effects. Furthermore, Cramp treatment reversed the oxidative stress by increasing the antioxidant enzymes of GSH-PX and decreasing the oxidant content of MDA. Notably, compared to the DSS group, Cramp treatment significantly elevated the abundance of Verrucomicrobiota at the phylum level. Furthermore, at the genus level, Parasutterella and Mucispirllum abundance was increased significantly in response to Cramp treatment, although Roseburia and Enterorhabdus reduced remarkably. Metabolic pathway analysis of serum metabolomics showed that Cramp intervention can regulate various metabolic pathways such as α-linolenic acid, taurine and hypotaurine, sphingolipid, and arachidonic acid metabolism. The study concluded that Cramp significantly ameliorated DSS-induced colonic injury, colonic inflammation, and intestinal barrier dysfunction in mice. The underlying mechanism is closely related to the metabolic alterations derived from gut microbiota.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Trauma Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Zhongyuan Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Haiyang Wang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Lichun Zhang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Mengjiao Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Gaoqian Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Chang Li
- Department of Trauma Center and Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Jilin, China
| | - Bo Wang
- Department of Trauma Center and Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Jilin, China
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| | - Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, Jilin, China
| |
Collapse
|
11
|
Wu J, Roesger S, Jones N, Hu CMJ, Li SD. Cell-penetrating peptides for transmucosal delivery of proteins. J Control Release 2024; 366:864-878. [PMID: 38272399 DOI: 10.1016/j.jconrel.2024.01.038] [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: 12/06/2023] [Revised: 01/11/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Enabling non-invasive delivery of proteins across the mucosal barriers promises improved patient compliance and therapeutic efficacies. Cell-penetrating peptides (CPPs) are emerging as a promising and versatile tool to enhance protein and peptide permeation across various mucosal barriers. This review examines the structural and physicochemical attributes of the nasal, buccal, sublingual, and oral mucosa that hamper macromolecular delivery. Recent development of CPPs for overcoming those mucosal barriers for protein delivery is summarized and analyzed. Perspectives regarding current challenges and future research directions towards improving non-invasive transmucosal delivery of macromolecules for ultimate clinical translation are discussed.
Collapse
Affiliation(s)
- Jiamin Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Sophie Roesger
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Natalie Jones
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| |
Collapse
|
12
|
Nakai D, Miyake M. Intestinal Membrane Function in Inflammatory Bowel Disease. Pharmaceutics 2023; 16:29. [PMID: 38258040 PMCID: PMC10820082 DOI: 10.3390/pharmaceutics16010029] [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: 10/31/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Inflammatory bowel disease is a set of chronic inflammatory diseases that mainly develop in the gastrointestinal mucosa, including ulcerative colitis and Crohn's disease. Gastrointestinal membrane permeability is an important factor influencing the pharmacological effects of pharmaceuticals administered orally for treating inflammatory bowel disease and other diseases. Understanding the presence or absence of changes in pharmacokinetic properties under a disease state facilitates effective pharmacotherapy. In this paper, we reviewed the gastrointestinal membrane function in ulcerative colitis and Crohn's disease from the perspective of in vitro membrane permeability and electrophysiological parameters. Information on in vivo permeability in humans is summarized. We also overviewed the inflammatory bowel disease research using gut-on-a-chip, in which some advances have recently been achieved. It is expected that these findings will be exploited for the development of therapeutic drugs for inflammatory bowel disease and the optimization of treatment options and regimens.
Collapse
Affiliation(s)
- Daisuke Nakai
- Drug Metabolism & Pharmacokinetics Research Laboratory, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masateru Miyake
- Pharmapack Co., Ltd., 1-27 Nakaokubo, Toyama 939-2243, Japan;
| |
Collapse
|
13
|
Elzayat H, Malik T, Al-Awadhi H, Taha M, Elghazali G, Al-Marzooq F. Deciphering salivary microbiome signature in Crohn's disease patients with different factors contributing to dysbiosis. Sci Rep 2023; 13:19198. [PMID: 37932491 PMCID: PMC10628307 DOI: 10.1038/s41598-023-46714-8] [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: 06/14/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease. An imbalanced microbiome (dysbiosis) can predispose to many diseases including CD. The role of oral dysbiosis in CD is poorly understood. We aimed to explore microbiome signature and dysbiosis of the salivary microbiome in CD patients, and correlate microbiota changes to the level of inflammation. Saliva samples were collected from healthy controls (HC) and CD patients (n = 40 per group). Salivary microbiome was analyzed by sequencing the entire 16S rRNA gene. Inflammatory biomarkers (C-reactive protein and calprotectin) were measured and correlated with microbiome diversity. Five dominant species were significantly enriched in CD, namely Veillonella dispar, Megasphaera stantonii, Prevotella jejuni, Dolosigranulum pigrum and Lactobacillus backii. Oral health had a significant impact on the microbiome since various significant features were cariogenic as Streptococcus mutans or periopathogenic such as Fusobacterium periodonticum. Furthermore, disease activity, duration and frequency of relapses impacted the oral microbiota. Treatment with monoclonal antibodies led to the emergence of a unique species called Simonsiella muelleri. Combining immunomodulatory agents with monoclonal antibodies significantly increased multiple pathogenic species such as Salmonella enterica, Escherichia coli, Klebsiella pneumoniae and Pseudomonas aeruginosa. Loss of diversity in CD was shown by multiple diversity indices. There was a significant negative correlation between gut inflammatory biomarkers (particularly calprotectin) and α-diversity, suggesting more inflammation associated with diversity loss in CD. Salivary dysbiosis was evident in CD patients, with unique microbiota signatures and perturbed species that can serve as disease biomarkers or potential targets for microbiota modulation. The interplay of various factors collectively contributed to dysbiosis, although each factor probably had a unique effect on the microbiome. The emergence of pathogenic bacteria in the oral cavity of CD patients is alarming since they can disturb gut homeostasis and induce inflammation by swallowing, or hematogenous spread of microbiota, their metabolites, or generated inflammatory mediators.
Collapse
Affiliation(s)
- Hala Elzayat
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE
| | - Talha Malik
- Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | - Haifa Al-Awadhi
- Department of Pediatric Gastroenterology, Tawam Hospital, Al Ain, UAE
| | - Mazen Taha
- Department of Internal Medicine, Tawam Hospital, Al Ain, UAE
| | - Gehad Elghazali
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE
- Department of Immunology, Sheikh Khalifa Medical City, Union71-Purehealth, Abu Dhabi, UAE
| | - Farah Al-Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, UAE.
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, UAE.
| |
Collapse
|
14
|
Dong G, Boothe K, He L, Shi Y, McCluskey LP. Altered peripheral taste function in a mouse model of inflammatory bowel disease. Sci Rep 2023; 13:18895. [PMID: 37919307 PMCID: PMC10622515 DOI: 10.1038/s41598-023-46244-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023] Open
Abstract
Increased sugar intake and taste dysfunction have been reported in patients with inflammatory bowel disease (IBD), a chronic disorder characterized by diarrhea, pain, weight loss and fatigue. It was previously unknown whether taste function changes in mouse models of IBD. Mice consumed dextran sodium sulfate (DSS) during three 7-day cycles to induce chronic colitis. DSS-treated mice displayed signs of disease, including significant weight loss, diarrhea, loss of colon architecture, and inflammation of the colon. After the last DSS cycle we assessed taste function by recording electrophysiological responses from the chorda tympani (CT) nerve, which transmits activity from lingual taste buds to the brain. DSS treatment significantly reduced neural taste responses to natural and artificial sweeteners. Responses to carbohydrate, salt, sour or bitter tastants were unaffected in mice with colitis, but umami responses were modestly elevated. DSS treatment modulated the expression of receptor subunits that transduce sweet and umami stimuli in oral taste buds as a substrate for functional changes. Dysregulated systemic cytokine responses or dysbiosis that occurs during chronic colitis may be upstream from changes in oral taste buds. We demonstrate for the first time that colitis alters taste input to the brain, which could exacerbate malnutrition in IBD patients.
Collapse
Affiliation(s)
- Guangkuo Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street/CA-3016, Augusta, GA, 30912, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Khaylie Boothe
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street/CA-3016, Augusta, GA, 30912, USA
| | - Lianying He
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street/CA-3016, Augusta, GA, 30912, USA
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street/CA-3016, Augusta, GA, 30912, USA
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Lynnette Phillips McCluskey
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street/CA-3016, Augusta, GA, 30912, USA.
| |
Collapse
|
15
|
Dong G, Boothe K, He L, Shi Y, McCluskey LP. Altered peripheral taste function in a mouse model of inflammatory bowel disease. RESEARCH SQUARE 2023:rs.3.rs-3304297. [PMID: 37720020 PMCID: PMC10503843 DOI: 10.21203/rs.3.rs-3304297/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Increased sugar intake and taste dysfunction have been reported in patients with inflammatory bowel disease (IBD), a chronic disorder characterized by diarrhea, pain, weight loss and fatigue. It was previously unknown whether taste function changes in mouse models of IBD. Mice consumed dextran sodium sulfate (DSS) during three 7-day cycles to induce chronic colitis. DSS-treated mice displayed signs of disease, including significant weight loss, diarrhea, loss of colon architecture, and inflammation of the colon. After the last DSS cycle we assessed taste function by recording electrophysiological responses from the chorda tympani (CT) nerve, which transmits activity from lingual taste buds to the brain. DSS treatment significantly reduced neural taste responses to natural and artificial sweeteners. Responses to carbohydrate, salt, sour or bitter tastants were unaffected in mice with colitis, but umami responses were modestly elevated. DSS treatment modulated the expression of receptor subunits that transduce sweet and umami stimuli in oral taste buds as a substrate for functional changes. Dysregulated systemic cytokine responses, or dysbiosis that occurs during chronic colitis may be upstream from changes in oral taste buds. We demonstrate for the first time that colitis alters taste input to the brain, which could exacerbate malnutrition in IBD patients.
Collapse
|