1
|
Chen M, Lin J, Miao D, Yang X, Feng M, Liu M, Xu L, Lin Q. The effect of preoperative mechanical bowel preparation in paediatric bowel surgery on postoperative wound related complications: A meta-analysis. Int Wound J 2024; 21:e14884. [PMID: 38654483 PMCID: PMC11040098 DOI: 10.1111/iwj.14884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/13/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Mechanical bowel preparation (MBP), a routine nursing procedure before paediatric bowel surgery, is widely should in clinical practice, but its necessity remains controversial. In a systematic review and meta-analysis, we evaluated the effect of preoperative MBP in paediatric bowel surgery on postoperative wound-related complications in order to analyse the clinical application value of MBP in paediatric bowel surgery. As of November 2023, we searched four online databases: the Cochrane Library, Embase, PubMed, and Web of Science. Two investigators screened the collected studies against inclusion and exclusion criteria, and ROBINS-I was used to evaluate the quality of studies. Using RevMan5.3, a meta-analysis of the collected data was performed, and a fixed-effect model or a random-effect model was used to analyse OR, 95% CI, SMD, and MD. A total of 11 studies with 2556 patients were included. Most of studies had moderate-to-severe quality bias. The results of meta-analysis showed no statistically significant difference in the incidence of complications related to postoperative infections in children with MBP before bowel surgery versus those with No MBP, wound infection (OR 1.11, 95% CI:0.76 ~ 1.61, p = 0.59, I2 = 5%), intra-abdominal infection (OR 1.26, 95% CI:0.58 ~ 2.77, p = 0.56, I2 = 9%). There was no significant difference in the risk of postoperative bowel anastomotic leak (OR 1.07, 95% CI:0.68 ~ 1.68, p = 0.78, I2 = 12%), and anastomotic dehiscence (OR 1.67, 95% CI:0.13 ~ 22.20, p = 0.70, I2 = 73%). Patients' intestinal obstruction did not show an advantage of undergoing MBP preoperatively, with an incidence of intestinal obstruction (OR 1.95, 95% CI:0.55 ~ 6.93, p = 0.30, I2 = 0%). Based on existing evidence that preoperative MBP in paediatric bowel surgery did not reduce the risk of postoperative wound complications, we cautiously assume that MBP before surgery is unnecessary for children undergoing elective bowel surgery. However, due to the limited number of study participants selected for this study and the overall low quality of evidence, the results need to be interpreted with caution. It is suggested that more high quality, large-sample, multicenter clinical trials are required to validate our findings.
Collapse
Affiliation(s)
- Meixue Chen
- Department of PediatricsDongguan Maternal and Child Health Care HospitalDongguanChina
| | - Jin Lin
- Chinese OphthalmologyJoint Shantou International Eye Center of Shantou University and The Chinese University of Hong KongShantouChina
| | - Dongrong Miao
- Department of PediatricsDongguan Maternal and Child Health Care HospitalDongguanChina
| | - Xin Yang
- Department of NursingJinan University Affiliated First HospitalGuangzhouChina
| | - Mei Feng
- Department of NursingJinan University Affiliated First HospitalGuangzhouChina
| | - Manli Liu
- Department of NursingJinan University Affiliated First HospitalGuangzhouChina
| | - Lianqing Xu
- Department of PediatricsDongguan Maternal and Child Health Care HospitalDongguanChina
| | - Qingran Lin
- Department of NursingJinan University Affiliated First HospitalGuangzhouChina
| |
Collapse
|
2
|
Martins RDS, Kooi EMW, Poelstra K, Hulscher JBF. The role of intestinal alkaline phosphatase in the development of necrotizing enterocolitis. Early Hum Dev 2023; 183:105797. [PMID: 37300991 DOI: 10.1016/j.earlhumdev.2023.105797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating neonatal disease that affects neonates worldwide and often leads to high morbidity and mortality rates. Despite extensive research, the cause of NEC remains unclear, and current treatment options are limited. An important novel finding is the potential role of intestinal Alkaline Phosphatase (IAP) in both pathogenesis and treatment of NEC. IAP can play a vital role in detoxifying liposaccharides (LPS), a key mediator of many pathological processes, thereby reducing the inflammatory response associated with NEC. Furthermore, IAP can help prevent dysbiosis, improve intestinal perfusion, and promote autophagy. In this comprehensive review, we present evidence of the possible connection between IAP and the LPS/Toll-like receptor 4 (TLR4) pathway, impaired gut immunity, and dysbiosis in the preterm gut. Based on these findings, the administration of exogenous IAP might provide promising preventive and therapeutic avenues for the management of NEC.
Collapse
Affiliation(s)
- Raquel Dos Santos Martins
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Elisabeth M W Kooi
- Division of Neonatology, Department of Pediatrics, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Klaas Poelstra
- Department of Nanomedicine and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, the Netherlands
| | - Jan B F Hulscher
- Division of Pediatric Surgery, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
3
|
Núñez-Sánchez MA, Herisson FM, Keane JM, García-González N, Rossini V, Pinhiero J, Daly J, Bustamante-Garrido M, Hueston CM, Patel S, Canela N, Herrero P, Claesson MJ, Melgar S, Nally K, Caplice NM, Gahan CG. Microbial bile salt hydrolase activity influences gene expression profiles and gastrointestinal maturation in infant mice. Gut Microbes 2022; 14:2149023. [PMID: 36420990 PMCID: PMC9704388 DOI: 10.1080/19490976.2022.2149023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The mechanisms by which early microbial colonizers of the neonate influence gut development are poorly understood. Bacterial bile salt hydrolase (BSH) acts as a putative colonization factor that influences bile acid signatures and microbe-host signaling pathways and we considered whether this activity can influence infant gut development. In silico analysis of the human neonatal gut metagenome confirmed that BSH enzyme sequences are present as early as one day postpartum. Gastrointestinal delivery of cloned BSH to immature gnotobiotic mice accelerated shortening of the colon and regularized gene expression profiles, with monocolonised mice more closely resembling conventionally raised animals. In situ expression of BSH decreased markers of cell proliferation (Ki67, Hes2 and Ascl2) and strongly increased expression of ALPI, a marker of cell differentiation and barrier function. These data suggest an evolutionary paradigm whereby microbial BSH activity potentially influences bacterial colonization and in-turn benefits host gastrointestinal maturation.
Collapse
Affiliation(s)
- María A. Núñez-Sánchez
- APC Microbiome Ireland, University College Cork, Cork, Ireland,Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
| | - Florence M. Herisson
- APC Microbiome Ireland, University College Cork, Cork, Ireland,Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
| | - Jonathan M. Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - Natalia García-González
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - Valerio Rossini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jorge Pinhiero
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - Jack Daly
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | | | - Cara M. Hueston
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Shriram Patel
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - Nuria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira I Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Reus, Spain
| | - Pol Herrero
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira I Virgili-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Reus, Spain
| | - Marcus J. Claesson
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken Nally
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Biochemistry & Cell Biology, University College Cork, Cork, Ireland
| | - Noel M. Caplice
- APC Microbiome Ireland, University College Cork, Cork, Ireland,Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
| | - Cormac G.M. Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Microbiology, University College Cork, Cork, Ireland,School of Pharmacy, University College Cork, Cork, Ireland,CONTACT Cormac G.M. Gahan APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
4
|
Hao X, Ding N, Zhang Y, Yang Y, Zhao Y, Zhao J, Li Y, Li Z. Benign regulation of the gut microbiota: The possible mechanism through which the beneficial effects of manual acupuncture on cognitive ability and intestinal mucosal barrier function occur in APP/PS1 mice. Front Neurosci 2022; 16:960026. [PMID: 35992924 PMCID: PMC9382294 DOI: 10.3389/fnins.2022.960026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
Background Gut microbiota dysbiosis and intestinal barrier injury play vital roles in Alzheimer's disease (AD) onset and development. Our previous studies have demonstrated that manual acupuncture (MA) could improve the cognitive abilities of APP/PS1 mice. However, the effect of MA on the intestinal mucosal barrier and the gut microbiota mechanism through which this effect occurs remain to be clarified. Methods In the APP/PS1 manual acupuncture (Am) group, MA was applied in Baihui (GV20), Yintang (GV29), and Zusanli (ST36). Mice in the APP/PS1 antibiotic + manual acupuncture (Aa) group were treated with an antibiotic mixture and MA at the same time. Probiotics were delivered to the APP/PS1 probiotics (Ap) group. Alterations in spatial learning and memory, the gut microbiota, the intestinal barrier function, and the expression of glial fibrillary acidic protein (GFAP), lipopolysaccharide (LPS), and TNF-α were evaluated in each group. Results Compared with the C57BL/6 control (Cc) group, cognitive ability was significantly decreased, the gut microbiota structure was obviously disrupted, intestinal barrier integrity was drastically impaired, and the intestinal inflammatory response was enhanced in the APP/PS1 control (Ac) group (P < 0.01). These changes were reversed by MA and probiotics (P < 0.01 or P < 0.05), whereas antibiotics inhibited the benign regulation by MA (P < 0.01 or P < 0.05). Conclusion Manual acupuncture can benignly modulate gut microbiota dysbiosis, significantly reduce intestinal inflammation, and effectively alleviate the destruction of the intestinal mucosal barrier in APP/PS1 mice, and the effects are comparable to those of probiotics. The gut microbiota may play an important role in the improvement of the cognitive function and intestinal barrier function by MA.
Collapse
Affiliation(s)
- Xin Hao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ning Ding
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yichen Yang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yali Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yiran Li
- International School, Beijing University of Chinese Medicine, Beijing, China
| | - Zhigang Li
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Wu H, Wang Y, Li H, Meng L, Zheng N, Wang J. Protective Effect of Alkaline Phosphatase Supplementation on Infant Health. Foods 2022; 11:foods11091212. [PMID: 35563935 PMCID: PMC9101100 DOI: 10.3390/foods11091212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 12/03/2022] Open
Abstract
Alkaline phosphatase (ALP) is abundant in raw milk. Because of its high heat resistance, ALP negative is used as an indicator of successful sterilization. However, pasteurized milk loses its immune protection against allergy. Clinically, ALP is also used as an indicator of organ diseases. When the activity of ALP in blood increases, it is considered that diseases occur in viscera and organs. Oral administration or injecting ALP will not cause harm to the body and has a variety of probiotic effects. For infants with low immunity, ALP intake is a good prebiotic for protecting the infant’s intestine from potential pathogenic bacteria. In addition, ALP has a variety of probiotic effects for any age group, including prevention and treatment intestinal diseases, allergies, hepatitis, acute kidney injury (AKI), diabetes, and even the prevention of aging. The prebiotic effects of alkaline phosphatase on the health of infants and consumers and the content of ALP in different mammalian raw milk are summarized. The review calls on consumers and manufacturers to pay more attention to ALP, especially for infants with incomplete immune development. ALP supplementation is conducive to the healthy growth of infants.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62816069
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
6
|
Kini A, Zhao B, Basic M, Roy U, Iljazovic A, Odak I, Ye Z, Riederer B, Di Stefano G, Römermann D, Koenecke C, Bleich A, Strowig T, Seidler U. Upregulation of antimicrobial peptide expression in slc26a3-/- mice with colonic dysbiosis and barrier defect. Gut Microbes 2022; 14:2041943. [PMID: 35230892 PMCID: PMC8890434 DOI: 10.1080/19490976.2022.2041943] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Genetic defects in SLC26A3 (DRA), an intestinal Cl-/HCO3- exchanger, result in congenital chloride diarrhea (CLD), marked by lifelong acidic diarrhea and a high risk of inflammatory bowel disease. Slc26a3-/- mice serve as a model to understand the pathophysiology of CLD and search for treatment options. This study investigates the microbiota changes in slc26a3-/- colon, the genotype-related causes for the observed microbiota alterations, its inflammatory potential, as well as the corresponding host responses. The luminal and the mucosa-adherent cecal and colonic microbiota of cohoused slc26a3-/- and wt littermates were analyzed by 16S rRNA gene sequencing. Fecal microbiota transfer from cohoused slc26a3-/- and wt littermates to germ-free wt mice was performed to analyze the stability and the inflammatory potential of the communities.The cecal and colonic luminal and mucosa-adherent microbiota of slc26a3-/- mice was abnormal from an early age, with a loss of diversity, of short-chain fatty acid producers, and an increase of pathobionts. The transfer of slc26a3-/- microbiota did not result in intestinal inflammation and the microbial diversity in the recipient mice normalized over time. A strong increase in the expression of Il22, Reg3β/γ, Relmβ, and other proteins with antimicrobial functions was observed in slc26a3-/- colon from juvenile age, while the mucosal and systemic inflammatory signature was surprisingly mild. The dysbiotic microbiota, low mucosal pH, and mucus barrier defect in slc26a3-/- colon are accompanied by a stark upregulation of the expression of a panel of antimicrobial proteins. This may explain the low inflammatory burden in the gut of these mice.
Collapse
Affiliation(s)
| | - Bei Zhao
- Microbial Immune Regulation Research Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Urmi Roy
- Microbial Immune Regulation Research Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Aida Iljazovic
- Microbial Immune Regulation Research Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Ivan Odak
- Institute of Immunology Hannover Medical School Hannover, Germany
| | | | | | | | | | | | | | - Till Strowig
- Microbial Immune Regulation Research Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | |
Collapse
|
7
|
Orso G, Solovyev MM, Facchiano S, Tyrikova E, Sateriale D, Kashinskaya E, Pagliarulo C, Hoseinifar HS, Simonov E, Varricchio E, Paolucci M, Imperatore R. Chestnut Shell Tannins: Effects on Intestinal Inflammation and Dysbiosis in Zebrafish. Animals (Basel) 2021; 11:ani11061538. [PMID: 34070355 PMCID: PMC8228309 DOI: 10.3390/ani11061538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary With the increase in global population the production of animal proteins becomes increasingly crucial. Aquaculture is the first animal protein supply industry for human consumption. Intensive farming techniques are employed to increase productivity, but these may cause stressful conditions for fish, resulting in impaired growth and poor health conditions. Intestinal inflammation is one of the most common diseases of fish in intensive farming. Intestinal inflammation is usually accompanied by an alteration of the microbiota or dysbiosis. Inflammation and dysbiosis are so tightly intertwined that inflammation may contribute to or result from dysregulation of gut microbiota. Natural substances of plant origin rich in bioactive molecules or more simply phytochemicals, have been proved to be able to reduce inflammation and improve the general health status in various commercially relevant species. In this study, we evaluated the effect of tannins, a class of polyphenols, the most abundant phytochemicals, on intestinal inflammation and microbiota in zebrafish (Danio rerio), a small freshwater fish become an attractive biomedicine and aquaculture animal model during the last decades. The zebrafish has been employed in a vast array of studies aiming at investigating the essential processes underlying intestinal inflammation and injury due to its conservative gut morphology and functions. In this study, we administered a diet enriched with chestnut shell extract rich in tannins to a zebrafish model of intestinal inflammation. The treatment ameliorated the damaged intestinal morphophysiology and the microbiota asset. Our results sustain that products of natural origin with low environmental impact and low cost, such as tannins, may help to ease some of the critical issues affecting the aquaculture sector. Abstract The aim of the present study was to test the possible ameliorative efficacy of phytochemicals such as tannins on intestinal inflammation and dysbiosis. The effect of a chestnut shell (Castanea sativa) extract (CSE) rich in polyphenols, mainly represented by tannins, on k-carrageenan-induced intestinal inflammation in adult zebrafish (Danio rerio) was tested in a feeding trial. Intestinal inflammation was induced by 0.1% k-carrageenan added to the diet for 10 days. CSE was administered for 10 days after k-carrageenan induced inflammation. The intestinal morphology and histopathology, cytokine expression, and microbiota were analyzed. The k-carrageenan treatment led to gut lumen expansion, reduction of intestinal folds, and increase of the goblet cells number, accompanied by the upregulation of pro-inflammatory factors (TNFα, COX2) and alteration in the number and ratio of taxonomic groups of bacteria. CSE counteracted the inflammatory status enhancing the growth of health helpful bacteria (Enterobacteriaceae and Pseudomonas), decreasing the pro-inflammatory factors, and activating the anti-inflammatory cytokine IL-10. In conclusion, CSE acted as a prebiotic on zebrafish gut microbiota, sustaining the use of tannins as food additives to ameliorate the intestinal inflammation. Our results may be relevant for both aquaculture and medical clinic fields.
Collapse
Affiliation(s)
- Graziella Orso
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| | - Mikhail M. Solovyev
- Institute of Systematics and Ecology of Animals, Siberian Branch of RAS, 630091 Novosibirsk, Russia; (M.M.S.); (E.T.); (E.K.)
- Biological Institute, Tomsk State University, 634050 Tomsk, Russia
| | - Serena Facchiano
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| | - Evgeniia Tyrikova
- Institute of Systematics and Ecology of Animals, Siberian Branch of RAS, 630091 Novosibirsk, Russia; (M.M.S.); (E.T.); (E.K.)
- Department of Natural Sciences, Novosibirsk State University, 630091 Novosibirsk, Russia
| | - Daniela Sateriale
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| | - Elena Kashinskaya
- Institute of Systematics and Ecology of Animals, Siberian Branch of RAS, 630091 Novosibirsk, Russia; (M.M.S.); (E.T.); (E.K.)
| | - Caterina Pagliarulo
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| | - Hossein S. Hoseinifar
- Department of Fisheries, Gorgan University of Agricultural Sciences and Natural Resources, 49138-15739 Gorgan, Iran;
| | - Evgeniy Simonov
- Institute of Environmental and Agricultural Biology (X-BIO), University of Tyumen, 625003 Tyumen, Russia;
| | - Ettore Varricchio
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| | - Marina Paolucci
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
- Correspondence:
| | - Roberta Imperatore
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy; (G.O.); (S.F.); (D.S.); (C.P.); (E.V.); (R.I.)
| |
Collapse
|
8
|
Alvarenga L, Cardozo LFMF, Lindholm B, Stenvinkel P, Mafra D. Intestinal alkaline phosphatase modulation by food components: predictive, preventive, and personalized strategies for novel treatment options in chronic kidney disease. EPMA J 2020; 11:565-579. [PMID: 33240450 PMCID: PMC7680467 DOI: 10.1007/s13167-020-00228-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Alkaline phosphatase (AP) is a ubiquitous membrane-bound glycoprotein that catalyzes phosphate monoesters' hydrolysis from organic compounds, an essential process in cell signaling. Four AP isozymes have been described in humans, placental AP, germ cell AP, tissue nonspecific AP, and intestinal AP (IAP). IAP plays a crucial role in gut microbial homeostasis, nutrient uptake, and local and systemic inflammation, and its dysfunction is associated with persistent inflammatory disorders. AP is a strong predictor of mortality in the general population and patients with cardiovascular and chronic kidney disease (CKD). However, little is known about IAP modulation and its possible consequences in CKD, a disease characterized by gut microbiota imbalance and persistent low-grade inflammation. Mitigating inflammation and dysbiosis can prevent cardiovascular complications in patients with CKD, and monitoring factors such as IAP can be useful for predicting those complications. Here, we review IAP's role and the results of nutritional interventions targeting IAP in experimental models to prevent alterations in the gut microbiota, which could be a possible target of predictive, preventive, personalized medicine (PPPM) to avoid CKD complications. Microbiota and some nutrients may activate IAP, which seems to have a beneficial impact on health; however, data on CKD remains scarce.
Collapse
Affiliation(s)
- L. Alvarenga
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
| | - L. F. M. F. Cardozo
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| | - B. Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - P. Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - D. Mafra
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| |
Collapse
|
9
|
Beneficial Effect of Mildly Pasteurized Whey Protein on Intestinal Integrity and Innate Defense in Preterm and Near-Term Piglets. Nutrients 2020; 12:nu12041125. [PMID: 32316586 PMCID: PMC7230795 DOI: 10.3390/nu12041125] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/08/2023] Open
Abstract
Background. The human digestive tract is structurally mature at birth, yet maturation of gut functions such as digestion and mucosal barrier continues for the next 1–2 years. Human milk and infant milk formulas (IMF) seem to impact maturation of these gut functions differently, which is at least partially related to high temperature processing of IMF causing loss of bioactive proteins and formation of advanced glycation end products (AGEs). Both loss of protein bioactivity and formation of AGEs depend on heating temperature and time. The aim of this study was to investigate the impact of mildly pasteurized whey protein concentrate (MP-WPC) compared to extensively heated WPC (EH-WPC) on gut maturation in a piglet model hypersensitive to enteral nutrition. Methods. WPC was obtained by cold filtration and mildly pasteurized (73 °C, 30 s) or extensively heat treated (73 °C, 30 s + 80 °C, 6 min). Preterm (~90% gestation) and near-term piglets (~96% gestation) received enteral nutrition based on MP-WPC or EH-WPC for five days. Macroscopic and histologic lesions in the gastro-intestinal tract were evaluated and intestinal responses were further assessed by RT-qPCR, immunohistochemistry and enzyme activity analysis. Results. A diet based on MP-WPC limited epithelial intestinal damage and improved colonic integrity compared to EH-WPC. MP-WPC dampened colonic IL1-β, IL-8 and TNF-α expression and lowered T-cell influx in both preterm and near-term piglets. Anti-microbial defense as measured by neutrophil influx in the colon was only observed in near-term piglets, correlated with histological damage and was reduced by MP-WPC. Moreover, MP-WPC stimulated iALP activity in the colonic epithelium and increased differentiation into enteroendocrine cells compared to EH-WPC. Conclusions. Compared to extensively heated WPC, a formula based on mildly pasteurized WPC limits gut inflammation and stimulates gut maturation in preterm and near-term piglets and might therefore also be beneficial for preterm and (near) term infants.
Collapse
|
10
|
Khan MS, Ikram M, Park JS, Park TJ, Kim MO. Gut Microbiota, Its Role in Induction of Alzheimer's Disease Pathology, and Possible Therapeutic Interventions: Special Focus on Anthocyanins. Cells 2020; 9:cells9040853. [PMID: 32244729 PMCID: PMC7226756 DOI: 10.3390/cells9040853] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
The human gut is a safe environment for several microbes that are symbiotic and important for the wellbeing of human health. However, studies on gut microbiota in different animals have suggested that changes in the composition and structure of these microbes may promote gut inflammation by releasing inflammatory cytokines and lipopolysaccharides, gut-wall leakage, and may affect systemic inflammatory and immune mechanisms that are important for the normal functioning of the body. There are many factors that aid in the gut’s dysbiosis and neuroinflammation, including high stress levels, lack of sleep, fatty and processed foods, and the prolonged use of antibiotics. These neurotoxic mechanisms of dysbiosis may increase susceptibility to Alzheimer’s disease (AD) and other neurodegenerative conditions. Therefore, studies have recently been conducted to tackle AD-like conditions by specifically targeting gut microbes that need further elucidation. It was suggested that gut dyshomeostasis may be regulated by using available options, including the use of flavonoids such as anthocyanins, and restriction of the use of high-fatty-acid-containing food. In this review, we summarize the gut microbiota, factors promoting it, and possible therapeutic interventions especially focused on the therapeutic potential of natural dietary polyflavonoid anthocyanins. Our study strongly suggests that gut dysbiosis and systemic inflammation are critically involved in the development of neurodegenerative disorders, and the natural intake of these flavonoids may provide new therapeutic opportunities for preclinical or clinical studies.
Collapse
Affiliation(s)
- Muhammad Sohail Khan
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.S.K.); (M.I.); (J.S.P.)
| | - Muhammad Ikram
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.S.K.); (M.I.); (J.S.P.)
| | - Jun Sung Park
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.S.K.); (M.I.); (J.S.P.)
| | - Tae Ju Park
- Paul O’Gorman Leukaemia Research, Centre Institute of Cancer, Sciences University of Glasgow, 0747 657 5394 Glasgow, UK;
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.S.K.); (M.I.); (J.S.P.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
11
|
Abstract
Background: Antibiotics are frequently used to treat critically ill patients, and its use is often accompanied by intestinal dysbiosis that might further lead to bacterial translocation (BT). Nevertheless, studies on the relationship between antibiotic therapy and BT are rare. In the present study, we investigated the effect of broad-spectrum antibiotics on BT in an experimental rat model of burn or sepsis injury. Methods: The septic rat model was simulated by a second insult with lipopolysaccharides after burn injury. Ninety-two male Sprague-Dawley rats were randomly divided into control, burn, and sepsis groups (n = 8 or 9, each group), and the latter two groups were then treated with imipenem or ceftriaxone for 3 or 9 days. The mesenteric lymph nodes, liver, lungs, and blood were collected at each time point under sterile conditions for quantitative bacterial culture and strain identification. The differences between the groups were compared by Fisher exact test or Mann-Whitney U test. Results: Only minimal Escherichia coli translocation to the mesenteric lymph nodes was observed in the normal control group, in which the BT rate was 12.5%. Burn injury did not affect the BT rate (Burn group vs. Control group, 12.5% vs. 12.5%, P = 1.000), whereas the BT rate showed an increased trend after the second insult with lipopolysaccharide (Sepsis group vs. Control group, 44.4% vs. 12.5%, P = 0.294), and many strains of Enterobacteria spp. were detected in distant organs (liver, lung, and blood) [Sepsis group vs. Control group, 0 (0,3) vs. 0 (0,0), U = 20, P = 0.045]. After the antibiotic treatment, BT to the distant organs was increased in burned rats [Burn IT3 group vs. Burn group, 0 (0,2) vs. 0 (0,0); Burn IT9 group vs. Burn group, 0 (0,1) vs. 0 (0,0); Burn CT9 group vs. Burn group, 0 (0,2) vs. 0 (0,0); all U = 20 and P = 0.076] but decreased in septic rats [Sepsis CT3 group vs. Sepsis group, 0 (0,0) vs. 0 (0,3), U = 20, P = 0.045]. The total amount of translocated bacteria, regardless of which antibiotic was used, was increased in burned rats [Burn IT9 group vs. Burn group, 2.389 (0,2.845) vs. 0 (0,2.301) Log10 colony-forming units (CFU)/g, U = 14, P = 0.034; Burn CT3 group vs. Burn group, 2.602 (0,3.633) vs. 0 (0,2.301) Log10 CFU/g, U = 10.5, P = 0.009], but there was a slightly decreased trend in septic rats [Sepsis IT9 group vs. Sepsis group, 2.301 (2,3.146) vs. 0 (0,4.185) Log10 CFU/g, U = 36, P = 0.721; Sepsis CT9 group vs. Sepsis group, 2 (0,3.279) vs. 0 (0,4.185) Log10 CFU/g, U = 32.5, P = 0.760]. Remarkably, the quantity of Enterococci spp. dramatically increased after broad-spectrum antibiotic treatment in both the burned and septic groups [Burn IT3 group vs. Burn group, 1 (0,5.164) vs. 0 (0,0) Log10 CFU/g, U = 16; Burn IT9 group vs. Burn group, 1 (0,2.845) vs. 0 (0,0) Log10 CFU/g, U = 16; Burn CT3 group vs. Burn group, 2.602 (0,3.633) vs. 0 (0,0) Log10 CFU/g, U = 8; Burn CT9 group vs. Burn group, 1 (0,4.326) vs. 0 (0,0) Log10 CFU/g, U = 16; Sepsis IT3 group vs. Sepsis group, 2.477 (0,2.903) vs. 0 (0,0) Log10 CFU/g, U = 4.5; Sepsis IT9 group vs. Sepsis group, 2 (0,3.146) vs. 0 (0,0) Log10 CFU/g, U = 9; Sepsis CT3 group vs. Sepsis group, 1.151 (0,2.477) vs. 0 (0,0) Log10 CFU/g, U = 18; Sepsis CT9 group vs. Sepsis group, 2 (0,3) vs. 0 (0,0) Log10 CFU/g, U = 13.5; all P < 0.05]. Conclusions: Broad-spectrum antibiotics promote BT in burned rats but prevent BT in septic rats, especially preventing BT to distant organs, such as the liver and lung. Moreover, Enterococci spp. with high drug resistance and high pathogenicity translocated most after antibiotic treatment.
Collapse
|
12
|
Wan J, Tian Z, Yao BY, Liu C, He JN, Yin X, Shi Y. Role of intestinal alkaline phosphatase in intestinal mucosal barrier. Shijie Huaren Xiaohua Zazhi 2019; 27:1441-1445. [DOI: 10.11569/wcjd.v27.i23.1441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intestinal alkaline phosphatase (IAP) is an alkaline phosphatase that plays an important role in maintaining the stability of the bowel function and the intestinal mucosal barrier, including adjusting the duodenal pH, participating the development of the intestinal tract, regulating the absorption ability of intestinal epithelial cells, reducing the toxicity of lipopolysaccharide, preventing and reducing the intestinal inflammation, regulating intestinal flora, improving intestinal calcium absorption, etc. In this paper, we will review the role of IAP in intestinal mucosal barrier.
Collapse
Affiliation(s)
- Jun Wan
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Zhong Tian
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Bai-Yu Yao
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Chong Liu
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Jing-Ni He
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Xin Yin
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Yang Shi
- the tenth department of surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
13
|
Arias-Jayo N, Abecia L, Lavín JL, Tueros I, Arranz S, Ramírez-García A, Pardo MA. Host-microbiome interactions in response to a high-saturated fat diet and fish-oil supplementation in zebrafish adult. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
14
|
Wang Y, Li X, Chen X, Zhao P, Qu Z, Ma D, Zhao C, Gao W. Effect of stir-frying time during Angelica Sinensis Radix processing with wine on physicochemical, structure properties and bioactivities of polysaccharides. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Abstract
PURPOSE OF REVIEW To summarize the current understanding and recent advances on the genetic aetiology in the pathogenesis of very early onset inflammatory bowel disease (VEO-IBD). RECENT FINDINGS IBD is a chronic disorder of the gastrointestinal tract whose manifestation is a result of complex interactions between genetics, environment, immune system and microbial flora. Over 230 IBD risk loci have been reported in genome wide association studies but the genetic contribution of the majority of these loci in the manifestation of IBD is very low. Patients with VEO-IBD present with a more severe disease than older patients, characterized by poor prognosis and failure of conventional therapy. Recent studies have reported several monogenic diseases with high penetrance that present with IBD and IBD-like intestinal manifestations and overlap with primary immunodeficiencies. Increasing body of evidence supports a prominent role of genetics in the onset of VEO-IBD. New genetic variants and diagnoses in VEO-IBD are reviewed and current challenges in therapy with potential strategy to manage the disease are discussed. SUMMARY Functional analysis of the genes implicated in monogenic IBD has increased the understanding of the underlying pathobiological mechanism of the disease. This knowledge can be used to personalize medicine for specific patients, improving the standard of care and quality of life.
Collapse
Affiliation(s)
- Vritika Batura
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Institute for Medical Science and Biochemistry, University of Toronto, Hospital for Sick Children,Toronto, Ontario, Canada
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Institute for Medical Science and Biochemistry, University of Toronto, Hospital for Sick Children,Toronto, Ontario, Canada
| |
Collapse
|
16
|
Arias-Jayo N, Abecia L, Alonso-Sáez L, Ramirez-Garcia A, Rodriguez A, Pardo MA. High-Fat Diet Consumption Induces Microbiota Dysbiosis and Intestinal Inflammation in Zebrafish. MICROBIAL ECOLOGY 2018; 76:1089-1101. [PMID: 29736898 DOI: 10.1007/s00248-018-1198-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/27/2018] [Indexed: 05/26/2023]
Abstract
Energy-dense foods and overnutrition represent major starting points altering lipid metabolism, systemic inflammation and gut microbiota. The aim of this work was to investigate the effects of a high-fat diet (HFD) over a period of 25 days on intestinal microbiota and inflammation in zebrafish. Microbial composition of HFD-fed animals was analysed and compared to controls by 16S rRNA sequencing and quantitative PCR. The expression level on several genes related to inflammation was tested. Furthermore, microscopic assessment of the intestine was performed in both conditions. The consumption of the HFD resulted in microbial dysbiosis, characterised by an increase in the relative abundance of the phylum Bacteroidetes. Moreover, an emerging intestinal inflammation via NF-κβ activation was confirmed by the overexpression of several genes related to signalling receptors, antimicrobial metabolism and the inflammatory cascade. The intestinal barrier was also damaged, with an increase of goblet cell mucin production. This is the first study performed in zebrafish which suggests that the consumption of a diet enriched with 10% fat changes the intestinal microbial community composition, which was correlated with low-grade inflammation.
Collapse
Affiliation(s)
- Nerea Arias-Jayo
- Food research, Azti, Parque tecnológico de Bizkaia, Astondo Bidea 609, 48160, Derio, Spain.
| | - Leticia Abecia
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Edificio 801A, 48160, Derio, Spain
| | - Laura Alonso-Sáez
- Marine research, Azti, Txatxarramendi ugartea z/g, 48395, Txatxarramendi, Spain
| | - Andoni Ramirez-Garcia
- Departmento de Immunología, Microbiología y Parasitología, Facultad de Ciencia y Tecnología, Universidad del País Vasco (UPV/EHU), 48940, Leioa, Spain
| | - Alfonso Rodriguez
- St Luke's General Hospital, Freshford Road, Friarsinch, Kilkenny, R95 FY71, Ireland
| | - Miguel A Pardo
- Food research, Azti, Parque tecnológico de Bizkaia, Astondo Bidea 609, 48160, Derio, Spain
| |
Collapse
|
17
|
Physicochemical characterizations of polysaccharides from Angelica Sinensis Radix under different drying methods for various applications. Int J Biol Macromol 2018; 121:381-389. [PMID: 30315881 DOI: 10.1016/j.ijbiomac.2018.10.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 01/31/2023]
Abstract
Polysaccharides (ASP) were extracted from Angelica Sinensis Radix (ASR) and were subjected to freeze drying (ASP-FD), vacuum drying (ASP-VD), and hot air drying (ASP-HD). Structural characteristics, rheological and emulsifying properties, antioxidant, anticoagulant and alkaline phosphatase inhibitory activities of ASPs were firstly investigated. ASP-FD owned higher thermostability, unique morphological structure, uniform and the lowest molecular weight, which is suitable for using as a raw material for polysaccharide derivatives. Smaller viscosity of ASP-FD supplied a better appearance that consumers preferred and the smallest particle size was benefit to manufacture instant soluble products. ASP-VD behaved the highest neutral sugar content, protein content, intrinsic viscosities, and the best emulsifying activity, being a candidate for cosmetics additive. ASP-HD exhibited the highest apparent viscosities, higher thermostability, loose structure, and better bioactivities. Drying conditions had significant effect on the physicochemical properties and bioactivities of ASPs. ASPs with specific characteristics may meet the requirements for diverse applications.
Collapse
|
18
|
Parlato M, Charbit-Henrion F, Pan J, Romano C, Duclaux-Loras R, Le Du MH, Warner N, Francalanci P, Bruneau J, Bras M, Zarhrate M, Bègue B, Guegan N, Rakotobe S, Kapel N, De Angelis P, Griffiths AM, Fiedler K, Crowley E, Ruemmele F, Muise AM, Cerf-Bensussan N. Human ALPI deficiency causes inflammatory bowel disease and highlights a key mechanism of gut homeostasis. EMBO Mol Med 2018; 10:emmm.201708483. [PMID: 29567797 PMCID: PMC5887907 DOI: 10.15252/emmm.201708483] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Herein, we report the first identification of biallelic-inherited mutations in ALPI as a Mendelian cause of inflammatory bowel disease in two unrelated patients. ALPI encodes for intestinal phosphatase alkaline, a brush border metalloenzyme that hydrolyses phosphate from the lipid A moiety of lipopolysaccharides and thereby drastically reduces Toll-like receptor 4 agonist activity. Prediction tools and structural modelling indicate that all mutations affect critical residues or inter-subunit interactions, and heterologous expression in HEK293T cells demonstrated that all ALPI mutations were loss of function. ALPI mutations impaired either stability or catalytic activity of ALPI and rendered it unable to detoxify lipopolysaccharide-dependent signalling. Furthermore, ALPI expression was reduced in patients' biopsies, and ALPI activity was undetectable in ALPI-deficient patient's stool. Our findings highlight the crucial role of ALPI in regulating host-microbiota interactions and restraining host inflammatory responses. These results indicate that ALPI mutations should be included in screening for monogenic causes of inflammatory bowel diseases and lay the groundwork for ALPI-based treatments in intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marianna Parlato
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN
| | - Fabienne Charbit-Henrion
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Department of Pediatric Gastroenterology, Assistance Publique-Hôpitaux de Paris Hôpital Necker-Enfants Malades, Paris, France
| | - Jie Pan
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Claudio Romano
- GENIUS group from ESPGHAN.,Unit of Pediatrics, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina, Italy
| | - Rémi Duclaux-Loras
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Marie-Helene Le Du
- Department of Biochemistry, Biophysics and Structural Biology, Institute for Integrative Biology of the Cell (I2BC), CEA, UMR 9198 CNRS, Université Paris-Sud, Gif-sur-Yvette, France
| | - Neil Warner
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Paola Francalanci
- Digestive Endoscopy and Surgery Unit and Pathology Unit Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Julie Bruneau
- Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Department of Pathology, Necker-Enfants Malades Hospital Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marc Bras
- Bioinformatics Platform, Université Paris-Descartes-Paris Sorbonne Centre and Institut Imagine, Paris, France
| | - Mohammed Zarhrate
- Genomic Platform, INSERM, UMR1163, Imagine Institute, Paris Descartes-Sorbonne Paris Cite University, Paris, France
| | - Bernadette Bègue
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN
| | - Nicolas Guegan
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Sabine Rakotobe
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN
| | - Nathalie Kapel
- Department of Functional Coprology, Pitié Salpêtrière Hospital Assistance publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit and Pathology Unit Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Anne M Griffiths
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Karoline Fiedler
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Eileen Crowley
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - Frank Ruemmele
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France.,GENIUS group from ESPGHAN.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Department of Pediatric Gastroenterology, Assistance Publique-Hôpitaux de Paris Hôpital Necker-Enfants Malades, Paris, France
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada .,Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Nadine Cerf-Bensussan
- INSERM, UMR1163, Laboratory of Intestinal Immunity and Institut Imagine, Paris, France .,GENIUS group from ESPGHAN.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| |
Collapse
|
19
|
Rader BA. Alkaline Phosphatase, an Unconventional Immune Protein. Front Immunol 2017; 8:897. [PMID: 28824625 PMCID: PMC5540973 DOI: 10.3389/fimmu.2017.00897] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Recent years have seen an increase in the number of studies focusing on alkaline phosphatases (APs), revealing an expanding complexity of function of these enzymes. Of the four human AP (hAP) proteins, most is known about tissue non-specific AP (TNAP) and intestinal AP (IAP). This review highlights current understanding of TNAP and IAP in relation to human health and disease. TNAP plays a role in multiple processes, including bone mineralization, vitamin B6 metabolism, and neurogenesis, is the genetic cause of hypophosphatasia, influences inflammation through regulation of purinergic signaling, and has been implicated in Alzheimer's disease. IAP regulates fatty acid absorption and has been implicated in the regulation of diet-induced obesity and metabolic syndrome. IAP and TNAP can dephosphorylate bacterial-derived lipopolysaccharide, and IAP has been identified as a potential regulator of the composition of the intestinal microbiome, an evolutionarily conserved function. Endogenous and recombinant bovine APs and recombinant hAPs are currently being explored for their potential as pharmacological agents to treat AP-associated diseases and mitigate multiple sources of inflammation. Continued research on these versatile proteins will undoubtedly provide insight into human pathophysiology, biochemistry, and the human holobiont.
Collapse
Affiliation(s)
- Bethany A Rader
- Department of Microbiology, Southern Illinois University, Carbondale, IL, United States
| |
Collapse
|