1
|
Bertens CA, Stoffel C, Crombie MB, Vahmani P, Penner GB. The effects of dietary cation-anion difference and dietary buffer for lactating dairy cattle under mild heat stress with night cooling. J Dairy Sci 2024:S0022-0302(24)01165-2. [PMID: 39343199 DOI: 10.3168/jds.2024-25225] [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: 05/28/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024]
Abstract
The objective of this study was to investigate the interactive effect of dietary cation-anion difference (DCAD) and dietary buffer supply on DMI, ruminal fermentation, milk and milk component yields, and gastrointestinal tract (GIT) permeability in lactating dairy cattle exposed to mild heat stress. Sixteen lactating Holstein cows, including 8 ruminally cannulated primiparous (80 ± 19.2 DIM) and 8 non-cannulated multiparous (136 ± 38.8 DIM) cows, were housed in a tie-stall barn programmed to maintain a temperature-humidity index (THI) between 68 and 72 from 0600 h to 1600 h followed by natural night cooling. The experimental design was a replicated 4 × 4 Latin rectangle (21-d periods) with a 2 × 2 factorial treatment arrangement. Diets contained a low DCAD (LD; 17.5 mEq/100g of DM) or high DCAD (HD; 39.6 mEq/100g of DM) adjusted using NH4Cl and Na-acetate, with low (LB; 0% CaMg(CO3)2) or high buffer (HB; 1% CaMg(CO3)2). In addition to measurement of feed intake, ruminal fermentation, and milk and milk component yields, a ruminal dose of Cr-EDTA and an equimolar abomasal dose of Co-EDTA were used to evaluate total and post-ruminal gastrointestinal tract permeability, respectively. Treatments had no effect on DMI, ruminal short-chain fatty acid concentrations, or ruminal pH. Feeding HD improved blood acid-base balance, increased urine volume by 4 ± 1.5 kg/d, and increased milk fat by 0.14 ± 0.044 percentage units and milk fat yield by 36.5 ± 16.71 g/d. HB reduced milk fat percentage by 0.11 ± 0.044 percentage units and had no effect on milk fat yield. The HB treatments reduced urinary excretion of Co by 27% and tended to reduce urinary Cr excretion by 10%. Across all treatments, 72% of the Cr recovery was represented by Co suggesting that much of the permeability responses were post-ruminal during mild heat stress. In conclusion, increasing DCAD through greater Na supply during mild heat stress improved blood acid-base balance and may increase milk fat yield. Dietary inclusion of CaMg(CO3)2 improved post-ruminal GIT barrier function despite a lack of low ruminal pH. As there appeared to be a limited interactive effect between DCAD and buffer, increased DCAD and provision of buffer seem to independently influence physiological and performance responses in lactating dairy cows exposed to mild heat with night cooling.
Collapse
Affiliation(s)
- C A Bertens
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8
| | - C Stoffel
- Papillon Agricultural Company and MIN-AD Inc., Easton, MD 21601
| | - M B Crombie
- Papillon Agricultural Company and MIN-AD Inc., Easton, MD 21601
| | - P Vahmani
- Department of Animal Science, UC Davis, Davis, CA, USA 95616-5270
| | - G B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8.
| |
Collapse
|
2
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Pinsino A, Braghieri L, Carey MR, Clerkin K, Sayer GT, Latif F, Koji T, Uriel N, Nandakumar R, Uhlemann AC, Colombo PC, Demmer RT. Alterations in the sarcopenia index are associated with inflammation, gut, and oral microbiota among heart failure, left ventricular assist device, and heart transplant patients. J Heart Lung Transplant 2024; 43:1395-1408. [PMID: 38744352 DOI: 10.1016/j.healun.2024.04.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/28/2024] [Accepted: 04/27/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Sarcopenia, characterized by loss of muscle mass and function, is prevalent in heart failure (HF) and predicts poor outcomes. We investigated alterations in sarcopenia index (SI), a surrogate for skeletal muscle mass, in HF, left ventricular assist device (LVAD), and heart transplant (HT), and assessed its relationship with inflammation and digestive tract (gut and oral) microbiota. METHODS We enrolled 460 HF, LVAD, and HT patients. Repeated measures pre/post-procedures were obtained prospectively in a subset of LVAD and HT patients. SI (serum creatinine/cystatin C) and inflammatory biomarkers (C-reactive protein, interleukin-6, tumor necrosis factor-alpha) were measured in 271 and 622 blood samples, respectively. Gut and saliva microbiota were assessed via 16S ribosomal ribonucleic acid sequencing among 335 stool and 341 saliva samples. Multivariable regression assessed the relationship between SI and (1) New York Heart Association class; (2) pre- versus post-LVAD or HT; and (3) biomarkers of inflammation and microbial diversity. RESULTS Median (interquartile range) natural logarithm (ln)-SI was -0.13 (-0.32, 0.05). Ln-SI decreased across worsening HF class, further declined at 1 month after LVAD and HT, and rebounded over time. Ln-SI was correlated with inflammation (r = -0.28, p < 0.01), gut (r = 0.28, p < 0.01), and oral microbial diversity (r = 0.24, p < 0.01). These associations remained significant after multivariable adjustment in the combined cohort but not for all individual cohorts. The presence of the gut taxa Roseburia inulinivorans was associated with increased SI. CONCLUSIONS SI levels decreased in symptomatic HF and remained decreased long-term after LVAD and HT. In the combined cohort, SI levels covaried with inflammation in a similar fashion and were significantly related to overall microbial (gut and oral) diversity, including specific taxa compositional changes.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York.
| | - Bruno Bohn
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Annamaria Ladanyi
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Alberto Pinsino
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Lorenzo Braghieri
- Division of Cardiovascular Medicine, Department of Cardiology, Cleveland Clinic, Cleveland, Ohio
| | - Matthew R Carey
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Kevin Clerkin
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Gabriel T Sayer
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Farhana Latif
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Takeda Koji
- Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Nir Uriel
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases and Microbiome and Pathogen Genomics Core, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Department of Cardiology, New York Presbyterian Hospital, Columbia University, New York, New York
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota; Division of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
3
|
Bertens CA, Seymour DJ, Penner GB. Validation of an in vivo dual permeability marker technique to characterize regional gastrointestinal tract permeability in mid lactation Holstein cows during short-term feed restriction. J Dairy Sci 2024:S0022-0302(24)01103-2. [PMID: 39218063 DOI: 10.3168/jds.2024-25142] [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: 05/10/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
This study evaluated the impact of short-term feed restriction in lactating dairy cows on regional permeability of the gastrointestinal tract (GIT), and the recovery of DMI, ruminal pH, and milk yield. In addition, sampling methods for a novel dual marker technique to characterize total GIT and post ruminal permeability were validated. Six ruminally cannulated lactating Holstein cows were blocked by parity (3 primiparous, 3 multiparous; 189 DIM ± 25.2) and enrolled in a crossover design. Experimental periods included a 5-d baseline phase (BASE), 5-d challenge phase (CHAL), and 2 weeks of recovery (REC1 and REC2). During CHAL cows received either 100% ad libitum feed intake (AL) or 40% of ad libitum feed intake (FR). To assess, total-tract and post-ruminal permeability, equimolar doses of Cr-EDTA and Co-EDTA were infused on d 3 of CHAL into the rumen and abomasum (0.369 mmol/kg BW). Following infusions, total urine and feces were collected every 8 h over 96 h, and blood samples were collected at h 0, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24, 32, 40, 48, and 64. The plasma area under the curve (AUC) for Cr and Co were calculated. By design, DMI for FR was reduced by 60% during CHAL and remained 19% lower than AL during REC1 but was not different from AL in REC2. Mean ruminal pH for FR was greatest during CHAL and the least during REC1, with no differences detected between AL and FR in REC2. The duration that pH was < 5.8 was the least for FR during CHAL and greatest during REC1 which were different from AL and were no longer different between treatments in REC2. Milk yield was the least for FR during CHAL and REC1 and no longer different from AL in REC2. Feed restriction reduced milk fat, protein, and lactose yields by 26, 31% and 31%, respectively. Plasma Cr AUC was 34% greater and Co AUC tended to be 35% greater for FR than AL on d 3 of CHAL. Urinary Cr recovery after 48-h was not affected by treatment; however, urinary Co recovery was 36% greater for FR than AL. Positive correlations between plasma AUC and urinary recovery for Cr and Co were detected. It was determined that blood samples collected at h 2, 8, 20, 40, and 48 could predict the total plasma Cr and Co AUC within 1.9% and 6.2%, respectively. In summary, short-term FR in lactating dairy cows increases permeability of the total GIT and may increase permeability of the post-ruminal regions with more than 60% of the permeability occurring post-ruminally. After FR, cows experienced low ruminal pH and a sustained reduction in milk yield. When utilizing Cr- and Co-EDTA to evaluate regional GIT permeability, plasma AUC can be used as an alternate to urinary Cr and Co excretion. In addition, blood samples collected at h 2, 8, 20, 40, and 48 result in adequate prediction accuracy, at least when comparing GIT permeability for lactating dairy cows exposed to AL and FR.
Collapse
Affiliation(s)
- C A Bertens
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8
| | - D J Seymour
- Trouw Nutrition R&D, P.O. Box 200, 5830 AE Boxmeer, the Netherlands; Centre for Nutrition Modelling, Department of Animal Biosciences, University of Guelph, ON, Canada N1G 2W1
| | - G B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8.
| |
Collapse
|
4
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
5
|
Silva BC, Godoi LA, Supapong C, Bitsie B, Valadares Filho SC, Schoonmaker JP. Effect of a molasses-based liquid supplement on gastrointestinal tract barrier function, inflammation, and performance of newly received feedlot cattle before and after a transport stress. J Anim Sci 2023; 101:skac295. [PMID: 36592757 PMCID: PMC9831108 DOI: 10.1093/jas/skac295] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/02/2022] [Indexed: 01/04/2023] Open
Abstract
The objective of this study was to determine the effect of a dry versus a molasses-based liquid supplement on ruminal butyrate concentration, gastrointestinal tract (GIT) barrier function, inflammatory status, and performance of newly received feedlot cattle. In experiment 1, 60 mixed breed steers (234 ± 2.1 kg) were weaned, held overnight at a sale barn, then transported 14 h to Purdue University. After arrival, steers were weighed, blocked by body weight, and allotted within block to treatments (six pens per treatment and five steers per pen). Diets consisted of 45% roughage and 55% concentrate (dry matter basis). Treatments differed in the supplement source as follows: DRY: 10% dry supplement or LIQUID: 10% liquid molasses-based supplement. Feed intake, average daily gain (ADG), and gain:feed were determined for the three 21-d periods and overall. In experiment 2, 16 crossbred heifers (246 ± 7.5 kg) were used (8 heifers per treatment). Diets were the same as in experiment 1 and were fed for 60 d. On d 56 ruminal fluid samples were collected at 0, 3, 6, and 9 h after feeding. To mimic a stress event, heifers were transported for 4 h on d 61, rested overnight, and transported 12 h on d 62. Blood was collected from heifers immediately prior to transport and immediately upon their return. Gut barrier function using a Cr-EDTA marker was determined after transportation. Data were analyzed using the GLIMMIX procedure of SAS. Steers fed the liquid supplement had greater (P ≤ 0.03) ADG through d 42 and overall compared to steers fed the dry supplement. Feed intake did not differ (P = 0.25) between treatments from d 0 to d 21. However, steers fed the liquid supplement showed greater (P < 0.001) dry matter intake after d 21 and overall compared to those fed the dry supplement. Steers fed the liquid supplement tended (P < 0.09) to have reduced serum haptoglobin and lipopolysaccharide-binding protein (LBP) compared to those fed the dry supplement. Heifers fed the liquid supplement had greater (P = 0.02) Cr in urine and tended (P = 0.07) to have lower serum LBP after transport compared to those fed the dry supplement. Heifers fed the liquid supplement had 72% lower serum haptoglobin before, but only a 19% lower serum haptoglobin after transport compared to animals fed the dry supplement (treatment × time; P = 0.07). Therefore, the liquid supplement altered GIT barrier function, and improved inflammatory status, resulting in increased growth of receiving cattle.
Collapse
Affiliation(s)
- Breno C Silva
- Department of Animal Sciences, Federal University of Viçosa, Vicosa, MG, Brazil
| | - Leticia A Godoi
- Department of Animal Sciences, Federal University of Viçosa, Vicosa, MG, Brazil
| | - Chanadol Supapong
- Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen, Khon Kaen Province, Thailand
| | - Bryce Bitsie
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | | | - Jon P Schoonmaker
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
6
|
El Mouzan M, Al-Hussaini AA, Al Sarkhy A, Assiri A, Alasmi M. Intestinal microbiota profile in healthy Saudi children: The bacterial domain. Saudi J Gastroenterol 2022; 28:312-317. [PMID: 35848701 PMCID: PMC9408733 DOI: 10.4103/sjg.sjg_585_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Knowledge of microbiota in health is essential for clinical research on the role of microbiota in disease. We aimed to characterize the intestinal microbiota in healthy Saudi children. Methods In this community-based study, stool samples were collected from a randomly selected sample of 20 healthy school children of Saudi origin. The samples were frozen at -80°C till analysis. Bacterial DNA was isolated and libraries were prepared using the Illumina Nextera XT library preparation kit. Unassembled sequencing reads were directly analyzed and quantified for each organism's relative abundance. The abundance for each organism was calculated and expressed as the average relative percentage from phyla to species. Results The median age was 11.3 (range 6.8-15.4) years, and 35% of them were males. The three most abundant phyla were Firmicutes, Bacteroidetes, and Actinobacteria accounting for 49%, 26%, and 24%, respectively. The most abundant genera included Bifidobacterium, Bacteroides, and Blautia accounting for 18.9%, 12.8%, and 8.2%, respectively. Finally, the most abundant species included 14 species belonging to the genus Bacteroides and nine species belonging to Bifidobacterium. Conclusions The abundance of intestinal microbiome in healthy Saudi children is different from that of other populations. Further studies are needed to understand the causes of variation between populations, which might lead to new preventive methods and treatment strategies of diseases caused by microbial dysbiosis.
Collapse
Affiliation(s)
- Mohammad El Mouzan
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulrahman A Al-Hussaini
- Division of Pediatric Gastroenterology, Children's Specialized Hospital, King Fahad Medical City; Faculty of Medicine, AlFaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Al Sarkhy
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Asaad Assiri
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mona Alasmi
- Department of Pediatrics, Gastroenterology Division, King Saud University Medical City, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Li S, Ma Y, Ye S, Guo R, Su Y, Du Q, Yin S, Xiao F. Ambient NO 2 exposure induced cardiotoxicity associated with gut microbiome dysregulation and glycerophospholipid metabolism disruption. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113583. [PMID: 35561545 DOI: 10.1016/j.ecoenv.2022.113583] [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: 03/07/2022] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
An average daily increase of 10 μg/m3 in NO2 concentrations could lead to an increased mortality in cardiovascular, cerebrovascular of 1.89%, 2.07%, but the mechanism by which NO2 contributes to cardiotoxicity is rarely reported. In order to assess the cardiotoxicity of NO2 inhalation (5 ppm), we firstly investigate the change of gut microbiota, serum metabonomics and cardiac proteome. Non-targeted LC-MS/MS metabonomics showed that NO2 stress could perturb the glycerophospholipid metabolism in the serum, which might destabilize the bilayer configuration of cardiac lipid membranes. Furthermore, we observed that NO2 inhalation caused augmented intercellular gap and inflammatory infiltration in the heart. Although 16 S rRNA gene amplification sequencing demonstrated that NO2 exposure did not influence the intestinal microbial abundance and diversity, but glycerophospholipid metabolism disruption might be finally reflected in gut microbiom dysregulation, such as Sphingomonas, Koribacter, Actinomarina and Bradyrhizobium Turicibacter, Rothia, Globicatella and Aerococcus. Proteome mining revealed that differentially expressed genes (DEGs) in the heart after NO2 stress were involved in necroptosis, mitophagy and ferroptosis. We further revealed that NO2 increased the number of cardiac mitochondria with depletion of cristae by regulating the expression of Mfn2 and Hsp70. This study indicating Mfn2-meidcated imbalanced mitochondrial dynamics as a potential mechanism after NO2-induced heart injury and suggesting microbiome dysregulation/glycerophospholipid metabolism exerts critical roles in cardiotoxicity caused by NO2.
Collapse
Affiliation(s)
- Siwen Li
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| | - Yu Ma
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Shuzi Ye
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Rong Guo
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Ying Su
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Qiaoyun Du
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Siyu Yin
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Fang Xiao
- Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
8
|
Zhao S, Feng P, Meng W, Jin W, Li X, Li X. Modulated Gut Microbiota for Potential COVID-19 Prevention and Treatment. Front Med (Lausanne) 2022; 9:811176. [PMID: 35308540 PMCID: PMC8927624 DOI: 10.3389/fmed.2022.811176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has gained global attention. SARS-CoV-2 identifies and invades human cells via angiotensin-converting enzyme 2 receptors, which is highly expressed both in lung tissues and intestinal epithelial cells. The existence of the gut-lung axis in disease could be profoundly important for both disease etiology and treatment. Furthermore, several studies reported that infected patients suffer from gastrointestinal symptoms. The gut microbiota has a noteworthy effect on the intestinal barrier and affects many aspects of human health, including immunity, metabolism, and the prevention of several diseases. This review highlights the function of the gut microbiota in the host's immune response, providing a novel potential strategy through the use of probiotics, gut microbiota metabolites, and dietary products to enhance the gut microbiota as a target for COVID-19 prevention and treatment.
Collapse
Affiliation(s)
- Shuai Zhao
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Pengya Feng
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wenbo Meng
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Weilin Jin
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xun Li
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiangkai Li
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Ogundepo S, Chiamaka AM, Olatinwo M, Adepoju D, Aladesanmi MT, Celestine UO, Ali KC, Umezinwa OJ, Olasore J, Alausa A. The role of diosgenin in crohn’s disease. CLINICAL PHYTOSCIENCE 2022. [DOI: 10.1186/s40816-022-00338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractInflammatory bowel disease (IBD) is a chronic idiopathic inflammation that can grossly affect the entire gastrointestinal tract (GIT) from the mouth to the anus. Crohn’s disease is the most known type of IBD and has been the focus of attention due to its increase in prevalence worldwide. Although the etiology is yet to be elucidated, recent studies have pointed out Crohn’s disease to arise from a complex interaction between environmental influences, genetic predisposition, and altered gut microbiota, resulting in dysregulated adaptive and innate responses. The presenting hallmarks of Crohn’s disease may include weight loss, nausea, vomiting, abdominal pain, diarrhea, fever, or chills. Treatment is usually done with many approved immunosuppressive drugs and surgery. However, a promising avenue from natural compounds is a safer therapy due to its safe natural active ingredients and the strong activity it shows in the treatment and management of diseases. Diosgenin, “a major biologically active natural steroidal sapogenin found in Chinese yam,” has been widely reported as a therapeutic agent in the treatment of various classes of disorders such as hyperlipidemia, inflammation, diabetes, cancer, infection, and immunoregulation. In this review, an analysis of literature data on diosgenin employed as a therapeutic agent for the treatment of Crohn’s disease is approached, to strengthen the scientific database and curtail the dreadful impact of Crohn’s disease.
Collapse
|
10
|
Maio ACD, Basile G, Iacopetta D, Catalano A, Ceramella J, Cafaro D, Saturnino C, Sinicropi MS. The significant role of nutraceutical compounds in ulcerative colitis treatment. Curr Med Chem 2021; 29:4216-4234. [PMID: 34961429 DOI: 10.2174/0929867329666211227121321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease (IBD) mainly affecting the colon and the rectum. Its main characters are represented by relapsing and remitting mucosal inflammation, starting in the rectum and typically extending continuously proximally through part or the entire colon. UC pathogenesis depends on multiple factors, such as genetic predisposition, defects in the epithelial barrier, dysregulated immune responses, and environmental causes. The most frequent symptoms are abdominal pain, weight loss, mucus discharge, bloody diarrhoea, incontinence, nocturnal defecations, fever, and anemia. Existing therapies for UC include 5-aminosalicylic acid (5-ASA) and its derivatives, steroids, immunosuppressants and biological drugs. However, limited efficacy and unwanted adverse effects hardly limit these strategies of treatment. In the last decades, research studies have been driven towards complementary and alternative medicines for the treatment of UC. Various nutraceuticals have exhibited promising results in modulating intestinal inflammation meanwhile improving symptoms. These compounds possess a wide spectrum of positive health effects evidenced by in vitro studies, characterized by their involvement in antioxidant defenses, cell proliferation, and gene expression. The present review analyzes the available data about the different types of nutraceuticals and their potential effectiveness as adjuvant therapy of IBD, with particular emphasis to UC.
Collapse
Affiliation(s)
- Azzurra Chiara De Maio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Giovanna Basile
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126 Bari, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Danilo Cafaro
- Proctology Surgery, Tropea Hospital, Vibo Valentia, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| |
Collapse
|
11
|
Briggs NG, Silva BC, Godoi LA, Schoonmaker JP. Effect of aspirin to intentionally induce leaky gut on performance, inflammation, and carcass characteristics of feedlot cattle. J Anim Sci 2021; 99:6422613. [PMID: 34741613 DOI: 10.1093/jas/skab328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/03/2021] [Indexed: 12/30/2022] Open
Abstract
The negative impacts of stress on gastrointestinal tract (GIT) barrier function can result in compromised animal growth and health. Aspirin is known to cause mucosal injury leading to increased gut permeability and tight junction damage and can be used as a model to study leaky gut in cattle. The objective of this study was to determine the long-term impact of aspirin-induced chronic leaky gut on cattle growth and carcass attributes. Two treatments were evaluated in two studies: control (no aspirin) or 0.25% of the diet dry matter (DM) aspirin fed daily. Diets consisted of 50% corn, 24% dried distillers grains, 20% corn silage, and 6% supplement on a DM basis. In experiment 1, sixteen Angus × Simmental heifers, allotted by body weight (BW) and breed composition, were fed diets for 154 d. On day 155, heifers were dosed with 1 liter of a 180-mM Cr-ethylenediaminetetraacetic acid solution using an esophageal tube and had urine collected every 1.5 to 3 h for 48 h for analysis of Cr as a measure of gut leakiness. In experiment 2, ninety-six Simmental × Angus steers (355.0 ± 14.8 kg) were allotted by BW and breed composition and fed treatment diets for 159 d. Weight was recorded monthly and serum was collected on day 159 and analyzed for lipopolysaccharide-binding protein (LBP), interleukin-6 (IL-6), serum amyloid A (SAA), haptoglobin, and aspartate aminotransferase (AST). Data were analyzed using the MIXED procedure of SAS. Heifers fed 0.25% aspirin in experiment 1 excreted more Cr into urine compared with heifers not fed aspirin (overall treatment effect, P = 0.01). In experiment 2, aspirin tended to increase serum LBP (P = 0.06) but had no effect on concentrations of IL-6, haptoglobin, SAA, or AST (P ≥ 0.25). Aspirin tended to decrease average daily gain (P = 0.10), decreased hot carcass weight and rib-eye area (P ≤ 0.05), and increased fat thickness, marbling score, and yield grade (P ≤ 0.02). Aspirin tended to increase kidney, pelvic, and heart fat percentage (P = 0.10) and had no effect on liver abscesses (P ≥ 0.80). This study indicates that leaky gut induced by long-term administration of aspirin has negative impacts on feedlot performance and carcass composition. The negative impact of aspirin-induced leaky gut on animal performance suggests that chronic leaky gut caused by other factors (subacute acidosis, stress) may be a significant problem for the feedlot industry.
Collapse
Affiliation(s)
- Nathan G Briggs
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | - Breno C Silva
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | - Letícia A Godoi
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | - Jon P Schoonmaker
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Wang Y, Lv X, Li X, Zhao J, Zhang K, Hao X, Liu K, Liu H. Protective Effect of Lactobacillus plantarum P8 on Growth Performance, Intestinal Health, and Microbiota in Eimeria-Infected Broilers. Front Microbiol 2021; 12:705758. [PMID: 34305875 PMCID: PMC8299996 DOI: 10.3389/fmicb.2021.705758] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022] Open
Abstract
Coccidiosis is one of the major parasitic diseases in the commercial broiler industry. Probiotics can protect poultry against Eimeria infection. However, the mechanisms are not fully known. Therefore, Lactobacillus plantarum P8 (P8) was used to investigate its anti-coccidial property and mechanism. Five hundred broilers were allocated to five treatments: control diet (NC), control diet + Eimeria infection (IC), control diet containing 1 × 107 cfu/g P8 + Eimeria infection (P8L), control diet containing 1 × 108 cfu/g P8 + Eimeria infection (P8H), and control diet + Eimeria infection + Diclazuril (DIC). At day 14, all treatments except NC were inoculated with sporulated oocysts. Results indicated that Eimeria infection increased the mortality and oocysts shedding, and declined the growth performance as well as the intestinal barrier in Eimeria-treated broilers. On the contrary, dietary supplementation of low level P8, high level P8 and DIC decreased the mortality and oocysts shedding, but improved the growth performance and intestinal barrier. The impaired intestinal morphology in the IC group was also improved by P8H and DIC treatments. Besides, the elevated oxidative stress and pro-inflammation in Eimeria-infected broilers were reduced by P8L, P8H, and DIC treatments. Metagenomic analysis indicated P8 altered the structure of the gut microbiota, and the alteration was more obvious at day 21 than day 42. Notably, IC also increased the abundances of Eimeriidae, Eimeria and Eimeria tenella at day 21, while P8L and DIC decreased the abundances. Correlation analysis revealed that bacteria in Eimeria-treated broilers positively correlated with the intestinal permeability, oxidative stress and inflammation, while bacteria in broilers receiving P8L and DIC negatively correlated with the aforementioned pathological indices. Functional prediction demonstrated that the metagenomes of Eimeria-infected broilers were involved in several diseases. But the metagenomes of P8L-treated broilers were involved in energy metabolism and replication repair. In conclusion, dietary P8 supplementation inhibited oocyst shedding and improved the growth performance as well as the intestinal health of broilers infected with Eimeria, which was closely related to the regulation of gut microbiota. Moreover, the effects of P8 may be more effective in the early infection of coccidia.
Collapse
Affiliation(s)
- Yang Wang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Xiaoguo Lv
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Xuemin Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Jinshan Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Kai Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Xiaojing Hao
- Qingdao Institute of Animal Science and Veterinary Medicine, Qingdao, China
| | - Kaidong Liu
- Qingdao Institute of Animal Science and Veterinary Medicine, Qingdao, China
| | - Huawei Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
13
|
Wang J, Li X, Wu X, Wang Z, Wu X, Wang S, Jing G, Yan T. Fecal Microbiota Transplantation as an Effective Treatment for Carbapenem-Resistant Klebsiella pneumoniae Infection in a Renal Transplant Patient. Infect Drug Resist 2021; 14:1805-1811. [PMID: 34017186 PMCID: PMC8131010 DOI: 10.2147/idr.s308308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/29/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND In renal transplant recipients, carbapenem-resistant Klebsiella pneumoniae (CRKP) infection is a common complication, and usually associated with severe clinical outcomes due to a lack of effective treatment. CASE PRESENTATION A 37-year-old woman with CRKP infection one month after kidney transplantation was involved in this study. Microbial characteristics of fecal samples from the patient were analyzed. Fecal microbiota transplantation (FMT) was performed for treating the CRKP infection. One week after FMT, the patient's urine and anal swab cultures returned negative for CRKP, and 17 days after FMT, the incision showed complete healing. Moreover, the patient had no symptoms of infection two months after FMT. Alpha diversity analyses showed that before FMT, the patient was associated with obviously lower species richness and diversity than the donor, which significantly increased at one week, three weeks and two months after FMT. Beta diversity analyses showed that though the patient's microbial community post-FMT still differed from the donor composition, their distances decreased visibly, especially at one week and three weeks after FMT. Obvious shift in microbial composition could be observed before and after FMT. The microbial composition of the patient post FMT resembled the donor composition. Relative abundance of genera such as Phascolarctobacterium and Lachnoclostridium increased after FMT, while the relative abundance of Klebsiella significantly decreased. CONCLUSION This study demonstrated the therapeutic effect of FMT on infections caused by CRKP for a renal transplant patient. Further studies are required to confirm the findings of this study.
Collapse
Affiliation(s)
- Junpeng Wang
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xin Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, People’s Republic of China
| | - Xiaoqiang Wu
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Zhiwei Wang
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Xuan Wu
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Shanmei Wang
- Department of Clinical Microbiology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Gaopeng Jing
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| | - Tianzhong Yan
- Department of Urology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
14
|
Mamic P, Chaikijurajai T, Tang WHW. Gut microbiome - A potential mediator of pathogenesis in heart failure and its comorbidities: State-of-the-art review. J Mol Cell Cardiol 2020; 152:105-117. [PMID: 33307092 DOI: 10.1016/j.yjmcc.2020.12.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
Gut microbiome (GMB) has been increasingly recognized as a contributor to development and progression of heart failure (HF), immune-mediated subtypes of cardiomyopathy (myocarditis and anthracycline-induced cardiotoxicity), response to certain cardiovascular drugs, and HF-related comorbidities, such as chronic kidney disease, cardiorenal syndrome, insulin resistance, malnutrition, and cardiac cachexia. Gut microbiome is also responsible for the "gut hypothesis" of HF, which explains the adverse effects of gut barrier dysfunction and translocation of GMB on the progression of HF. Furthermore, accumulating evidence has suggested that gut microbial metabolites, including short chain fatty acids, trimethylamine N-oxide (TMAO), amino acid metabolites, and bile acids, are mechanistically linked to pathogenesis of HF, and could, therefore, serve as potential therapeutic targets for HF. Even though there are a variety of proposed therapeutic approaches, such as dietary modifications, prebiotics, probiotics, TMAO synthesis inhibitors, and fecal microbial transplant, targeting GMB in HF is still in its infancy and, indeed, requires further preclinical and clinical evidence. In this review, we aim to highlight the role gut microbiome plays in HF pathophysiology and its potential as a novel therapeutic target in HF.
Collapse
Affiliation(s)
- Petra Mamic
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, Stanford, CA, United States of America
| | - Thanat Chaikijurajai
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - W H Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States of America.
| |
Collapse
|
15
|
Briggs NG, Brennan KM, Funnell BJ, Nicholls GT, Schoonmaker JP. Use of aspirin to intentionally induce gastrointestinal tract barrier dysfunction in feedlot cattle. J Anim Sci 2020; 98:5894892. [PMID: 32815992 DOI: 10.1093/jas/skaa264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/12/2020] [Indexed: 11/12/2022] Open
Abstract
Stress negatively affects the gastrointestinal tract (GIT) barrier function, resulting in compromised animal health. A deeper understanding of how diet and stress impacts the GIT barrier function in feedlot cattle is needed. Aspirin decreases mucus production and mucosal repair in the GIT and could be used as a model for GIT barrier dysfunction research. The objective of this study was to evaluate the effectiveness of aspirin to induce GIT barrier dysfunction in beef cattle. In experiment 1, sixteen crossbred heifers (425.0 ± 8.6 kg) were allotted to 0, 50, 100, or 200 mg/kg body weight (BW) aspirin doses based on BW. Experiment 1 consisted of two periods separated by 4 wk where four heifers per treatment received the same aspirin dose during each period. Heifers were fed a 49.4% corn silage and 50.6% concentrate diet. The 200 mg/kg BW aspirin treatment was dosed as a 100 mg/kg BW aspirin oral bolus 36 and 24 h prior to Cr-ethylenediaminetetraacetic acid (EDTA) dosing (1 liter; 180 mM). The 50 and 100 mg/kg BW aspirin treatments were dosed as an oral bolus 24 h prior to Cr-EDTA dosing. Urine was collected every 3 h for 48 h and analyzed for Cr. Serum was collected at 0 and 48 h and analyzed for lipopolysaccharide-binding protein (LBP), interleukin-6, serum amyloid A (SAA), haptoglobin, and aspartate aminotransferase. In experiment 2, sixteen crossbred steers (576.0 ± 14.2 kg) fed a similar diet were allotted by BW to the 0 and 200 mg/kg BW aspirin treatments (eight steers/treatment) and were slaughtered 24 h after the last dose. Jejunal tissues were collected, and claudin (CLDN) 1, 2, and 3, occludin, and zonula occludens tight junction messenger ribonucleic acid (mRNA) expression was determined. Data were analyzed using the MIXED procedure of SAS. Urinary Cr excretion increased linearly at hours 3, 6, 9, and 12 (P ≤ 0.04) as aspirin dose increased from 0 to 200 mg/kg. Aspirin linearly increased Cr absorption (P = 0.02) and elimination (P = 0.04) rates and linearly decreased mean retention time of Cr (P = 0.02). Aspirin increased SAA (P = 0.04) and tended to increase LBP (P = 0.09) in serum but did not affect any other serum inflammatory marker (P ≥ 0.19). Aspirin tended to increase jejunal CLDN-1 mRNA expression (P = 0.10) but did not affect the mRNA expression of other genes regulating tight junction function (P ≥ 0.20). Results from this study indicate that aspirin disrupts the GIT barrier function in beef cattle and has a potential as a model in GIT permeability research.
Collapse
Affiliation(s)
- Nathan G Briggs
- Department of Animal Science, Purdue University, West Lafayette, IN
| | | | - Bethany J Funnell
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN
| | | | | |
Collapse
|
16
|
Markowiak P, Śliżewska K. The role of probiotics, prebiotics and synbiotics in animal nutrition. Gut Pathog 2018; 10:21. [PMID: 29930711 PMCID: PMC5989473 DOI: 10.1186/s13099-018-0250-0] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/26/2018] [Indexed: 12/15/2022] Open
Abstract
Along with the intensive development of methods of livestock breeding, breeders' expectations are growing concerning feed additives that would guarantee such results as accelerating growth rate, protection of health from pathogenic infections and improvement of other production parameters such as: absorption of feed and quality of meat, milk, eggs. The main reason for their application would be a strive to achieve some beneficial effects comparable to those of antibiotic-based growth stimulators, banned on 01 January 2006. High hopes are being associated with the use of probiotics, prebiotics and synbiotics. Used mainly for maintenance of the equilibrium of the intestinal microbiota of livestock, they turn out to be an effective method in fight against pathogens posing a threat for both animals and consumers. This paper discusses definitions of probiotics, prebiotics and synbiotics. Criteria that have to be met by those kinds of formulas are also presented. The paper offers a list of the most commonly used probiotics and prebiotics and some examples of their combinations in synbiotic formulas used in animal feeding. Examples of available study results on the effect of probiotics, prebiotics and synbiotics on animal health are also summarised.
Collapse
Affiliation(s)
- Paulina Markowiak
- Department of Biotechnology and Food Sciences, Institute of Fermentation Technology and Microbiology, Lodz University of Technology, ul. Wólczańska 171/173, 90-924 Lodz, Poland
| | - Katarzyna Śliżewska
- Department of Biotechnology and Food Sciences, Institute of Fermentation Technology and Microbiology, Lodz University of Technology, ul. Wólczańska 171/173, 90-924 Lodz, Poland
| |
Collapse
|
17
|
Gupta V, Nag D, Garg P. Recurrent urinary tract infections in women: How promising is the use of probiotics? Indian J Med Microbiol 2018; 35:347-354. [PMID: 29063878 DOI: 10.4103/ijmm.ijmm_16_292] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Urinary tract infections (UTIs) currently rank amongst the most prevalent bacterial infections, representing a major health hazard. UTIs in females usually start as vaginal infections and ascend to the urethra and bladder. Recurrent UTIs (rUTIs) can be defined as at least three episodes of UTI in 1 year or two episodes in 6 months. Various antibiotics have been the mainstay of therapy in ameliorating the incidence of UTIs, but recurrent infections continue to afflict many women. It necessitates the exploitation of alternative antimicrobial therapy. Probiotics have been shown to be effective in varied clinical trials for long-term preventions of rUTI. Because Escherichia coli is the primary pathogen involved in UTIs which spreads from the rectum to vagina and then ascends up the sterile urinary tract, improving the gut or vaginal flora will thus impact the urinary tract. Since a healthy vaginal microbiota is mainly dominated by Lactobacillus species, in this context, exogenously administered probiotics containing Lactobacilli play a pivotal role in reducing the risk of rUTI. The concept of artificially boosting the Lactobacilli numbers through probiotic administration has long been conceived but has been recently shown to be possible. Lactobacilli may especially be useful for women with a history of recurrent, complicated UTIs or on prolonged antibiotic use. Probiotics do not cause antibiotic resistance and may offer other health benefits due to vaginal re-colonisation with Lactobacilli. However, more comprehensive research is still needed, to recommend for probiotics as an alternative to antibiotics.
Collapse
Affiliation(s)
- Varsha Gupta
- Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| | - Deepika Nag
- Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| | - Pratibha Garg
- Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| |
Collapse
|
18
|
Nawaz A, Bakhsh Javaid A, Irshad S, Hoseinifar SH, Xiong H. The functionality of prebiotics as immunostimulant: Evidences from trials on terrestrial and aquatic animals. FISH & SHELLFISH IMMUNOLOGY 2018; 76:272-278. [PMID: 29510254 DOI: 10.1016/j.fsi.2018.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/21/2018] [Accepted: 03/02/2018] [Indexed: 05/24/2023]
Abstract
The gut immune system is, the main option for maintaining host's health, affected by numerous factors comprising dietary constituents and commensal bacteria. These dietary components that affect the intestinal immunity and considered as an alternative of antibiotics are called immunosaccharides. Fructooligosaccharide (FOS), Galactooligosaccharide (GOS), inulin, dietary carbohydrates, and xylooligosaccharide (XOS) are among the most studied prebiotics in human as well as in aquaculture. Although prebiotics and probiotics have revealed potential as treatment for numerous illnesses in both human and fish, a comprehensive understanding of the molecular mechanism behind direct and indirect effect on the intestinal immune response will help more and perhaps extra effective therapy intended for ailments. This review covers the most newly deep-rooted scientific outcomes about the direct and indirect mechanism through which these dietetic strategies can affect intestinal immunity of terrestrial and aquatic animals. Prebiotics exert an influence on gut immune system via the increase in lysozyme and phagocytic activity, macrophage activation and stimulation of monocyte-derived dendritic cells. Furthermore, these functional molecules also enhance epithelial barrier function, beneficial gut microbial population, and production of intermediate metabolites for example short chain fatty acids (SCFAs) that assist in balancing the immune system. Moreover, emphasis will be sited on the relationship among food/feed, the microbiota, and the gut immune system. In conclusion, further studies are nonetheless essential to confirm the direct effect of prebiotics on immune response.
Collapse
Affiliation(s)
- Asad Nawaz
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Allah Bakhsh Javaid
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Sana Irshad
- School of Environmental Studies, China University of Geosciences, Wuhan 430070, China
| | - Seyed Hossein Hoseinifar
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Hanguo Xiong
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
19
|
Cukrowska B, Sowińska A, Bierła JB, Czarnowska E, Rybak A, Grzybowska-Chlebowczyk U. Intestinal epithelium, intraepithelial lymphocytes and the gut microbiota - Key players in the pathogenesis of celiac disease. World J Gastroenterol 2017; 23:7505-7518. [PMID: 29204051 PMCID: PMC5698244 DOI: 10.3748/wjg.v23.i42.7505] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/31/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023] Open
Abstract
Celiac disease (CD) is a chronic immune-mediated disorder triggered by the ingestion of gluten in genetically predisposed individuals. Before activating the immune system, gluten peptides are transferred by the epithelial barrier to the mucosal lamina propria, where they are deamidated by intestinal tissue transglutaminase 2. As a result, they strongly bind to human leucocyte antigens (HLAs), especially HLA-DQ2 and HLA-DQ8, expressed on antigen-presenting cells. This induces an inflammatory response, which results in small bowel enteropathy. Although gluten is the main external trigger activating both innate and adaptive (specific) immunity, its presence in the intestinal lumen does not fully explain CD pathogenesis. It has been hypothesized that an early disruption of the gut barrier in genetically susceptible individuals, which would result in an increased intestinal permeability, could precede the onset of gluten-induced immune events. The intestinal barrier is a complex functional structure, whose functioning is dependent on intestinal microbiota homeostasis, epithelial layer integrity, and the gut-associated lymphoid tissue with its intraepithelial lymphocytes (IELs). The aim of this paper was to review the current literature and summarize the role of the gut microbiota, epithelial cells and their intercellular junctions, and IELs in CD development.
Collapse
Affiliation(s)
- Bożena Cukrowska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Agnieszka Sowińska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Joanna Beata Bierła
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children’s Memorial Health Institute, Warsaw 04-730, Poland
| | - Anna Rybak
- Department of Gastroenterology, Division of Neurogastroenterology and Motility, Great Ormond Street Hospital, London WC1N 3JH, United Kingdom
| | | |
Collapse
|
20
|
Hill DR, Huang S, Nagy MS, Yadagiri VK, Fields C, Mukherjee D, Bons B, Dedhia PH, Chin AM, Tsai YH, Thodla S, Schmidt TM, Walk S, Young VB, Spence JR. Bacterial colonization stimulates a complex physiological response in the immature human intestinal epithelium. eLife 2017; 6:29132. [PMID: 29110754 PMCID: PMC5711377 DOI: 10.7554/elife.29132] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/29/2017] [Indexed: 12/19/2022] Open
Abstract
The human gastrointestinal tract is immature at birth, yet must adapt to dramatic changes such as oral nutrition and microbial colonization. The confluence of these factors can lead to severe inflammatory disease in premature infants; however, investigating complex environment-host interactions is difficult due to limited access to immature human tissue. Here, we demonstrate that the epithelium of human pluripotent stem-cell-derived human intestinal organoids is globally similar to the immature human epithelium and we utilize HIOs to investigate complex host-microbe interactions in this naive epithelium. Our findings demonstrate that the immature epithelium is intrinsically capable of establishing a stable host-microbe symbiosis. Microbial colonization leads to complex contact and hypoxia driven responses resulting in increased antimicrobial peptide production, maturation of the mucus layer, and improved barrier function. These studies lay the groundwork for an improved mechanistic understanding of how colonization influences development of the immature human intestine.
Collapse
Affiliation(s)
- David R Hill
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Sha Huang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Melinda S Nagy
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Veda K Yadagiri
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Courtney Fields
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Dishari Mukherjee
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, United States
| | - Brooke Bons
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Priya H Dedhia
- Department of Surgery, University of Michigan, Ann Arbor, United States
| | - Alana M Chin
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Yu-Hwai Tsai
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Shrikar Thodla
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, United States
| | - Seth Walk
- Department of Microbiology and Immunology, Montana State University, Bozeman, United States
| | - Vincent B Young
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, United States
| | - Jason R Spence
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, United States.,Department of Cell andDevelopmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
21
|
Mϋller MJ, Paul T, Seeliger S. Necrotizing enterocolitis in premature infants and newborns. J Neonatal Perinatal Med 2017; 9:233-42. [PMID: 27589549 DOI: 10.3233/npm-16915130] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Necrotizing enterocolitis (NEC) is the most common acquired disease of the gastrointestinal tract (GIT) in premature infants and newborns. It is defined as an ulcerative inflammation of the intestinal wall. The clinical signs of incipient NEC are often very discrete, and range from localized intestinal symptoms to generalized signs of sepsis. NEC is classified depending on its severity into disease states according to the modified Bell's Classification. Treatment of NEC ranges, depending on its severity, from a conservative therapeutic approach to surgery with resection of the affected parts of the intestine. Mortality is considerably high in extremely small preterm infants reaching up to 42% of the affected children. Measures such as breastfeeding or alternatively nutrition with pasteurized human donor milk from a milk bank, administration of probiotics, avoidance of histamine type II receptor antagonists, and restrictive antibiotic treatment should be considered early on for prevention of NEC.
Collapse
Affiliation(s)
- M J Mϋller
- Department of Pediatric Cardiology and Intensive Care Medicine, Medical Center Georg August University Göttingen, Germany
| | - T Paul
- Department of Pediatric Cardiology and Intensive Care Medicine, Medical Center Georg August University Göttingen, Germany
| | - S Seeliger
- Department of Pediatric Cardiology and Intensive Care Medicine, Medical Center Georg August University Göttingen, Germany.,St. Elisabeth Children's Hospital, Neuburg/Donau, Germany
| |
Collapse
|
22
|
Peng Z, Vogel RF, Ehrmann MA, Xiong T. Identification and characterization of adhesion proteins in lactobacilli targeting actin as receptor. Mol Cell Probes 2017; 37:60-63. [PMID: 28823562 DOI: 10.1016/j.mcp.2017.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 10/19/2022]
Abstract
Actin as the main constitution of cytoskeleton in host cells plays an important role in mediating bacterial colonization. To identify the actin-binding proteins in Lactobacillus (L.) paracasei, L. plantarum, and L. brevis, actin immobilized to 24-well plate was used to probe adhesion proteins. Five adhesion proteins were identified and characterized by electrophoresis and LC-MS/MS: pyruvate kinase (PK), glucose-6-phosphate isomerase (PGI), phosphoglycerate kinase (PGK), chaperonin GroEL, and EF-Tu, all of which could display on the cell surface, indicating their possible role in mediating bacterial adhesion to host. This is in accordance with previous studies, which reported that these five proteins participated in and promoted the adhesion of pathogen or lactic acid bacteria to host. Moreover, PGK-actin binding domain analysis reveals that lysine (K) at amino acid position 127 in PGK might play a key role in mediating bacterial attachment to actin.
Collapse
Affiliation(s)
- Zhen Peng
- School of Food Science and Technology, Nanchang University, China; Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany; State Key Laboratory of Food Science and Technology, Nanchang University, China
| | - Rudi F Vogel
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany.
| | - Matthias A Ehrmann
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Freising, Germany
| | - Tao Xiong
- School of Food Science and Technology, Nanchang University, China; State Key Laboratory of Food Science and Technology, Nanchang University, China
| |
Collapse
|
23
|
Liao SF, Nyachoti M. Using probiotics to improve swine gut health and nutrient utilization. ACTA ACUST UNITED AC 2017; 3:331-343. [PMID: 29767089 PMCID: PMC5941265 DOI: 10.1016/j.aninu.2017.06.007] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 04/03/2017] [Accepted: 06/30/2017] [Indexed: 01/10/2023]
Abstract
To maintain a healthy gut is definitely key for a pig to digest and absorb dietary nutrients efficiently. A balanced microbiota (i.e., a healthy micro-ecosystem) is an indispensable constituent of a healthy gut. Probiotics, the live microorganisms which, when administered in adequate amounts, confer good health benefits onto the host, are a category of feed additives that can be used to replenish the gut microbial population while recuperating the host immune system. Besides their antitoxin and diarrhea reduction effects, dietary supplementation of probiotics can improve gut health, nutrient digestibilities and, therefore, benefit nutrient utilization and growth performance of pigs. Current knowledge in the literature pertinent to the beneficial effects of utilizing various probiotics for swine production has been comprehensively reviewed, and the safety and the risk issues related to probiotic usage have also been discussed in this paper. Considering that the foremost cost in a swine operation is feed cost, feed efficiency holds a very special, if not the paramount, significance in commercial swine production. Globally, the swine industry along with other animal industries is moving towards restricting and eventually a total ban on the usage of antibiotic growth promoters. Therefore, selection of an ideal alternative to the in-feed antibiotics to compensate for the lost benefits due to the ban on the antibiotic usage is urgently needed to support the industry for profitable and sustainable swine production. As is understood, a decision on this selection is not easy to make. Thus, this review paper aims to provide some much needed up-to-date knowledge and comprehensive references for swine nutritionists and producers to refer to before making prudent decisions and for scientists and researchers to develop better commercial products.
Collapse
Affiliation(s)
- Shengfa F Liao
- Department of Animal and Dairy Sciences, Mississippi State University, MS 39762, USA
| | - Martin Nyachoti
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
24
|
Sturgeon C, Fasano A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers 2016; 4:e1251384. [PMID: 28123927 DOI: 10.1080/21688370.2016.1251384] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/14/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
Beside digesting nutrients and absorbing solutes and electrolytes, the intestinal epithelium with its barrier function is in charge of a tightly controlled antigen trafficking from the intestinal lumen to the submucosa. This trafficking dictates the delicate balance between tolerance and immune response causing inflammation. Loss of barrier function secondary to upregulation of zonulin, the only known physiological modulator of intercellular tight junctions, leads to uncontrolled influx of dietary and microbial antigens. Additional insights on zonulin mechanism of action and the recent appreciation of the role that altered intestinal permeability can play in the development and progression of chronic inflammatory disorders has increased interest of both basic scientists and clinicians on the potential role of zonulin in the pathogenesis of these diseases. This review focuses on the recent research implicating zonulin as a master regulator of intestinal permeability linked to the development of several chronic inflammatory disorders.
Collapse
Affiliation(s)
- Craig Sturgeon
- Center for Celiac Research and Treatment, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Division of Pediatric Gastroenterology and Nutrition, Boston, MA, USA; Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alessio Fasano
- Center for Celiac Research and Treatment, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Division of Pediatric Gastroenterology and Nutrition, Boston, MA, USA; European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| |
Collapse
|
25
|
Uranga JA, López-Miranda V, Lombó F, Abalo R. Food, nutrients and nutraceuticals affecting the course of inflammatory bowel disease. Pharmacol Rep 2016; 68:816-26. [PMID: 27267792 DOI: 10.1016/j.pharep.2016.05.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/07/2016] [Accepted: 05/09/2016] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel diseases (ulcerative colitis; Crohn's disease) are debilitating relapsing inflammatory disorders affecting the gastrointestinal tract, with deleterious effect on quality of life, and increasing incidence and prevalence. Mucosal inflammation, due to altered microbiota, increased intestinal permeability and immune system dysfunction underlies the symptoms and may be caused in susceptible individuals by different factors (or a combination of them), including dietary habits and components. In this review we describe the influence of the Western diet, obesity, and different nutraceuticals/functional foods (bioactive peptides, phytochemicals, omega 3-polyunsaturated fatty acids, vitamin D, probiotics and prebiotics) on the course of IBD, and provide some hints that could be useful for nutritional guidance. Hopefully, research will soon offer enough reliable data to slow down the spread of the disease and to make diet a cornerstone in IBD therapy.
Collapse
Affiliation(s)
- José Antonio Uranga
- Área de Histología y Anatomía Patológica, Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Madrid, Spain; Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación (CIAL) del Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL). Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Madrid, Spain
| | - Visitación López-Miranda
- Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación (CIAL) del Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL). Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Madrid, Spain; Área de Farmacología y Nutrición, Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, URJC, Madrid, Spain; Unidad Asociada I+D+i al Instituto de Química Médica (IQM) del CSIC, Madrid, Spain
| | - Felipe Lombó
- Grupo de Investigación "Biotecnología de Nutracéuticos y Compuestos Bioactivos-BIONUC", Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Raquel Abalo
- Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación (CIAL) del Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL). Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos (URJC), Madrid, Spain; Área de Farmacología y Nutrición, Depto. de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, URJC, Madrid, Spain; Unidad Asociada I+D+i al Instituto de Química Médica (IQM) del CSIC, Madrid, Spain.
| |
Collapse
|
26
|
Pal PK, Hasan KN, Maitra SK. Gut melatonin response to microbial infection in carp Catla catla. FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:579-592. [PMID: 26563281 DOI: 10.1007/s10695-015-0161-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 11/03/2015] [Indexed: 06/05/2023]
Abstract
The purpose of present study was to demonstrate the response of gut melatoninergic system to Aeromonas hydrophila infection for 3 or 6 days and search for its correlation with the activity of different antioxidative and digestive enzymes to focus their interplay under pathophysiological conditions in carp (Catla catla). Microscopic study of gut in infected fish revealed degenerative changes in the tunica mucosa and lamina propria layers with sloughed off epithelial cells in the lumen. The activity of each digestive enzyme was reduced, but the levels of melatonin, arylalkylamine-N-acetyl transferase protein, the key regulator of melatonin biosynthesis, and different enzymatic antioxidants in gut were gradually and significantly increased with the progress of infection. Gut melatonin concentrations in A. hydrophila challenged carp by showing a positive correlation with the activity of each antioxidative enzyme, and a negative correlation with different digestive enzymes argued in favor of their functional relation, at least, during pathological stress. Moreover, parallel changes in the gut and serum melatonin titers indicated possible contribution of gut to circulating melatonin. Collectively, present carp study provided the first data to suggest that endogenous gut melatonin may be implicated to the mechanism of response to microbial infections in any fish species.
Collapse
Affiliation(s)
- Palash Kumar Pal
- Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Kazi Nurul Hasan
- Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Saumen Kumar Maitra
- Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India.
| |
Collapse
|
27
|
Dimitrellou D, Sidira M, Charalampopoulos D, Ypsilantis P, Galanis A, Simopoulos C, Kourkoutas Y. Effect of Cell Immobilization on Properties of Presumptive Probiotics. FOOD ENGINEERING SERIES 2016:257-268. [DOI: 10.1007/978-3-319-24040-4_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Abstract
Bacterial translocation is the invasion of indigenous intestinal bacteria through the gut mucosa to normally sterile tissues and the internal organs. Sometimes instead of bacteria, inflammatory compounds are responsible for clinical symptoms as in systemic inflammatory response syndrome (SIRS). The difference between sepsis and SIRS is that pathogenic bacteria are isolated from patients with sepsis but not with those of SIRS. Bacterial translocation occurs more frequently in patients with intestinal obstruction and in immunocompromised patients and is the cause of subsequent sepsis. Factors that can trigger bacterial translocation from the gut are host immune deficiencies and immunosuppression, disturbances in normal ecological balance of gut, mucosal barrier permeability, obstructive jaundice, stress, etc. Bacterial translocation occurs through the transcellular and the paracellular pathways and can be measured both directly by culture of mesenteric lymph nodes and indirectly by using labeled bacteria, peripheral blood culture, detection of microbial DNA or endotoxin and urinary excretion of non-metabolisable sugars. Bacterial translocation may be a normal phenomenon occurring on frequent basis in healthy individuals without any deleterious consequences. But when the immune system is challenged extensively, it breaks down and results in septic complications at different sites away from the main focus. The factors released from the gut and carried in the mesenteric lymphatics but not in the portal blood are enough to cause multi-organ failure. Thus, bacterial translocation may be a promoter of sepsis but not the initiator. This paper reviews literature on the translocation of gut flora and its role in causing sepsis.
Collapse
Affiliation(s)
- C Vaishnavi
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
29
|
Patel S, Shukla R, Goyal A. Probiotics in valorization of innate immunity across various animal models. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.02.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
30
|
Abstract
The field of genomics has expanded into subspecialties such as metagenomics over the course of the last decade and a half. The development of massively parallel sequencing capabilities has allowed for increasingly detailed study of the genome of the human microbiome, the microbial super organ that resides symbiotically within the mucosal tissues and integumentary system of the human host. The gut microbiome, and particularly the study of its origins in neonates, has become subtopics of great interest within the field of genomics. This brief review seeks to summarize recent literature regarding the origins and establishment of the neonatal gut microbiome, beginning in utero, and how it is affected by neonatal nutritional status (breastfed versus formula fed) and gestational age (term versus preterm). We also explore the role of dysbiosis, a perturbation within the fragile ecosystem of the microbiome, and its role in the origin of select pathologic states, specifically, obesity and necrotizing enterocolitis (NEC) in preterm infants. We discuss the evidence supporting enteral pre- and pro-biotic supplementation of commensal organisms such as Bifidobacterium and Lactobacillus in the neonatal period, and their role in the prevention and amelioration of NEC in premature infants. Finally, we review directions to consider for further research to promote human health within this field.
Collapse
Affiliation(s)
- Emily C Gritz
- Division of Perinatal Medicine, Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine , New Haven, CT , USA
| | - Vineet Bhandari
- Division of Perinatal Medicine, Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine , New Haven, CT , USA
| |
Collapse
|
31
|
Basic and clinical research on the regulation of the intestinal barrier by Lactobacillus and its active protein components: a review with experience of one center. Mol Biol Rep 2014; 41:8037-46. [PMID: 25185994 DOI: 10.1007/s11033-014-3701-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/23/2014] [Indexed: 12/15/2022]
Abstract
Probiotics got protective effects on the intestinal barrier. Our present study is to review the basic and clinical progress on the regulation of the intestinal barrier by Lactobacillus and its active protein components, combing the study of our center. Our study have isolated the active component of micro integral membrane protein (MIMP) within the media place of the integral membrane protein of Lactobacillus plantarum, which was verified about the protective effects against the intestinal epithelial dysfunction. On the other hand, we also found the effects of perioperative use of probiotics in the prevention and treatment of postoperative intestinal barrier dysfunction, and reduction of the postoperative infective complications. In this review, we would like to report the founding of our center, involving in the basic and clinical research progress of regulation of intestinal barrier by Lactobacillus and its active protein component MIMP. Furthermore, we may also promote our following studies about the MIMP and its clinical verification.
Collapse
|
32
|
Mercadante ACT, Perobelli SM, Alves APG, Gonçalves-Silva T, Mello W, Gomes-Santos AC, Miyoshi A, Azevedo V, Faria AMC, Bonomo A. Oral combined therapy with probiotics and alloantigen induces B cell-dependent long-lasting specific tolerance. THE JOURNAL OF IMMUNOLOGY 2014; 192:1928-37. [PMID: 24453248 DOI: 10.4049/jimmunol.1301034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allogeneic hematopietic stem cell transplantation (aHSCT) is widely used for the treatment of hematologic malignancies. Although aHSCT provides a good response against the malignant cells (graft-versus-leukemia [GVL]), it also leads to the development of graft-versus-host disease (GVHD), a severe disease with high mortality and morbidity rates. Therapy for GVHD is commonly based on nonspecific immunosupression of the transplanted recipient, resulting in the concomitant inhibition of the GVL effect. In this study, we propose an alternative approach to specifically suppress GVHD while sparing the GVL, based on oral treatment of transplant donors with recipient Ags, associated with the intake of probiotic Lactococcus lactis as tolerogenic adjuvant (combined therapy). We show that treatment of C57BL/6 donor mice with combined therapy before the transplant protects the recipients F1 (C57BL/6 × BAL/c) mice from clinical and pathological manifestations of disease, resulting in 100% survival rate. Importantly, the animals keep the immunological competence maintaining the GVL response as well as the response to third-party Ags. The protection is specific, long lasting and dependent on donor IL-10-sufficient B cells activity, which induces regulatory T cells in the host. These data suggest that combined therapy is a promising strategy for prevention of GVHD with preservation of GVL, opening new possibilities to treat human patients subjected to transplantation.
Collapse
Affiliation(s)
- Ana C T Mercadante
- Department of Experimental Medicine, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Toft-Hansen H, Nielsen C, Biagini M, Husby S, Lillevang ST. Lectin staining shows no evidence of involvement of glycocalyx/mucous layer carbohydrate structures in development of celiac disease. Nutrients 2013; 5:4540-52. [PMID: 24253051 PMCID: PMC3847747 DOI: 10.3390/nu5114540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/29/2013] [Accepted: 11/06/2013] [Indexed: 02/06/2023] Open
Abstract
The presence of unique carbohydrate structures in the glycocalyx/mucous layer of the intestine may be involved in a susceptibility to celiac disease (CD) by serving as attachment sites for bacteria. This host-microbiota interaction may influence the development of CD and possibly other diseases with autoimmune components. We examined duodenal biopsies from a total of 30 children, of which 10 had both celiac disease (CD) and type 1 diabetes (T1D); 10 had CD alone; and 10 were suspected of having gastrointestinal disease, but had normal duodenal histology (non-CD controls). Patients with both CD and T1D were examined before and after remission following a gluten-free diet. We performed lectin histochemistry using peanut agglutinin (PNA) and Ulex europaeus agglutinin (UEA) staining for Gal-β(1,3)-GalNAc and Fucα1-2Gal-R, respectively, of the glycocalyx/mucous layer. The staining was scored based on dissemination of stained structures on a scale from 0 to 3. Evaluation of the scores revealed no difference between biopsies obtained before and after remission in the group of children with both CD and T1D. A comparison of this pre-remission group with the children who had CD alone or the non-CD controls also showed no significant differences. Based on our material, we found no indication that the presence of Gal-β(1,3)-GalNAc or Fucα1-2Gal-R is involved in the susceptibility to CD, or that the disease process affects the expression of these carbohydrates.
Collapse
Affiliation(s)
- Henrik Toft-Hansen
- Hans Christian Andersen Children’s Hospital, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark; E-Mail:
- Department of Clinical Immunology, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark; E-Mails: (C.N.); (S.T.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +45-6541-1269; Fax: +45-6591-1862
| | - Christian Nielsen
- Department of Clinical Immunology, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark; E-Mails: (C.N.); (S.T.L.)
| | - Matteo Biagini
- Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 2., Odense 5000, Denmark; E-Mail:
| | - Steffen Husby
- Hans Christian Andersen Children’s Hospital, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark; E-Mail:
| | - Søren T. Lillevang
- Department of Clinical Immunology, Odense University Hospital, Sdr. Boulevard 29, Odense 5000, Denmark; E-Mails: (C.N.); (S.T.L.)
| |
Collapse
|
34
|
Fukuda S, Ohno H. Gut microbiome and metabolic diseases. Semin Immunopathol 2013; 36:103-14. [PMID: 24196453 DOI: 10.1007/s00281-013-0399-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/13/2013] [Indexed: 02/06/2023]
Abstract
The prevalence of obesity and obesity-related disorders is increasing worldwide. In the last decade, the gut microbiota has emerged as an important factor in the development of obesity and metabolic syndrome, through its interactions with dietary, environmental, and host genetic factors. Various studies have shown that alteration of the gut microbiota, shifting it toward increased energy harvest, is associated with an obese phenotype. However, the molecular mechanisms by which the gut microbiota affects host metabolism are still obscure. In this review, we discuss the complexity of the gut microbiota and its relationship to obesity and obesity-related diseases. Furthermore, we discuss the anti-obesity potential of probiotics and prebiotics.
Collapse
Affiliation(s)
- Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata, 997-0052, Japan
| | | |
Collapse
|
35
|
Chong ESL. A potential role of probiotics in colorectal cancer prevention: review of possible mechanisms of action. World J Microbiol Biotechnol 2013; 30:351-74. [PMID: 24068536 DOI: 10.1007/s11274-013-1499-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
A number of investigations, mainly using in vitro and animal models, have demonstrated a wide range of possible mechanisms, by which probiotics may play a role in colorectal cancer (CRC) prevention. In this context, the most well studied probiotics are certain strains from the genera of lactobacilli and bifidobacteria. The reported anti-CRC mechanisms of probiotics encompass intraluminal, systemic, and direct effects on intestinal mucosa. Intraluminal effects detailed in this review include competitive exclusion of pathogenic intestinal flora, alteration of intestinal microflora enzyme activity, reduction of carcinogenic secondary bile acids, binding of carcinogens and mutagens, and increasing short chain fatty acids production. Reduction of DNA damage and suppression of aberrant crypt foci formation have been well demonstrated as direct anti-CRC effects of probiotics on intestinal mucosa. Existing evidence clearly support a multifaceted immunomodulatory role of probiotics in CRC, particularly its ability to modulate intestinal inflammation, a well known risk factor for CRC. The effectiveness of probiotics in CRC prevention is dependent on the strain of the microorganism, while viability may not be a prerequisite for certain probiotic anticancer mechanisms, as indicated by several studies. Emerging data suggest synbiotic as a more effective approach than either prebiotics or probiotics alone. More in vivo especially human studies are warranted to further elucidate and confirm the potential role of probiotics (viable and non-viable), prebiotics and synbiotics in CRC chemoprevention.
Collapse
Affiliation(s)
- Esther Swee Lan Chong
- Institute of Food, Nutrition and Human Health, Massey University, PO Box 11222, Palmerston North, 4442, New Zealand,
| |
Collapse
|
36
|
Nakayama T, Takeuchi D, Matsumura T, Akeda Y, Fujinaga Y, Oishi K. Alcohol consumption promotes the intestinal translocation of Streptococcus suis infections. Microb Pathog 2013; 65:14-20. [PMID: 24036179 DOI: 10.1016/j.micpath.2013.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/28/2013] [Accepted: 08/30/2013] [Indexed: 12/29/2022]
Abstract
Streptococcus suis is an emerging zoonotic agent. This study aimed to investigate whether S. suis is likely to translocate across the intestines of human hosts who have liver disease and/or consume alcohol. Both the alcoholism and cirrhosis models exhibited high mRNA expression of TGF and collagen1, but only the cirrhosis model had fibrosis in the liver. After both models were infected with S. suis, significantly different concentrations of S. suis were detected in the blood and brains of the alcoholism model (Blood: 36.4%; Brain: 31.8%) and the cirrhosis model (Blood: 62.5%; Brain: 62.5%) compared to the concentrations in the healthy mice (Blood: 15.4%; Brain: 0%). Trans-epithelial electrical resistance (TER) was used to examine the Caco-2 cells in the in vitro that had an S. suis infection combined with 1% ethanol. Although the ethanol did not influence the Caco-2 cells' barriers, it did rapidly decrease the barriers' TER value and then their E-cadherin compared to the infected Caco-2 cells without the ethanol treatment. Immunofluorescence also indicated that the barriers of the Caco-2 cells treated with ethanol were disrupted and that S. suis translocated from the apical to the basolateral side. This study demonstrated that alcohol consumption helped S. suis to translocate.
Collapse
Affiliation(s)
- T Nakayama
- Laboratory for Clinical Research on Infectious Disease, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Zhang S, Albornoz RI, Aschenbach JR, Barreda DR, Penner G. Short-term feed restriction impairs the absorptive function of the reticulo-rumen and total tract barrier function in beef cattle1. J Anim Sci 2013; 91:1685-95. [DOI: 10.2527/jas.2012-5669] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- S. Zhang
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - R. I. Albornoz
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - J. R. Aschenbach
- Institute of Veterinary Physiology, Free University of Berlin, D-14163 Berlin, Germany
| | - D. R. Barreda
- Department of Agriculture, Food and Nutritional Science, University of Alberta, AB T6G 2P5, Canada
| | - G.B. Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| |
Collapse
|
38
|
The role of cell surface architecture of lactobacilli in host-microbe interactions in the gastrointestinal tract. Mediators Inflamm 2013; 2013:237921. [PMID: 23576850 PMCID: PMC3610365 DOI: 10.1155/2013/237921] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 02/11/2013] [Indexed: 11/18/2022] Open
Abstract
Lactobacillus species can exert health promoting effects in the gastrointestinal tract (GIT) through many mechanisms, which include pathogen inhibition, maintenance of microbial balance, immunomodulation, and enhancement of the epithelial barrier function. Different species of the genus Lactobacillus can evoke different responses in the host, and not all strains of the same species can be considered beneficial. Strain variations may be related to diversity of the cell surface architecture of lactobacilli and the bacteria's ability to express certain surface components or secrete specific compounds in response to the host environment. Lactobacilli are known to modify their surface structures in response to stress factors such as bile and low pH, and these adaptations may help their survival in the face of harsh environmental conditions encountered in the GIT. In recent years, multiple cell surface-associated molecules have been implicated in the adherence of lactobacilli to the GIT lining, immunomodulation, and protective effects on intestinal epithelial barrier function. Identification of the relevant bacterial ligands and their host receptors is imperative for a better understanding of the mechanisms through which lactobacilli exert their beneficial effects on human health.
Collapse
|
39
|
Lee YK. Effects of diet on gut microbiota profile and the implications for health and disease. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2013; 32:1-12. [PMID: 24936357 PMCID: PMC4034294 DOI: 10.12938/bmfh.32.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/10/2012] [Indexed: 12/11/2022]
Abstract
Gut microbes are present in large concentrations on the human intestinal mucosal surface
and play important roles in health and disease of the host. Numerous groups of gut
microbes are associated with immunological and metabolic diseases and in maintaining
health status of the host. Among these health- and disease-associated gut microbes,
Bacteroides, Clostridium and Bifidobacterium appear
regularly in the list. Scientific and clinical evidence available to date indicates that
diet is a major driving factor for the establishment of the gut microbiome. Slow
digestible carbohydrates (human milk glycan, inulin and fructooligosaccharide), insoluble
complex carbohydrates and protein diets favor the growth of Bacteroides,
Clostridium and Bifidobacterium. Fat on the other hand
suppresses the number of Bacteroides, Clostridium and
Bifidobacterium; whereas polyphenols in general suppress
Bacteroides and Clodtridium but enhance the
Bifodobacterium. The implication is that dietary habits could be a
major determinant of health and disease susceptibility. Dietary strategies could be an
effective means of potentially inducing changes in intestinal microbiota and are certainly
achievable, thus facilitating correction of intestinal microbiome aberrations or
imbalances to improve our health. Most of the physiological and functional interactions
between individual dietary components and the concoction of foods in a meal and gut
microbiota have not yet been well studied. A concerted effort is required to acquire
better understanding of their interaction in order to rationally maintain our intestinal
microbiome homeostasis and general health through dietary intervention.
Collapse
Affiliation(s)
- Yuan-Kun Lee
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, 117597 Singapore
| |
Collapse
|
40
|
Zhang HL, Yu LX, Yang W, Tang L, Lin Y, Wu H, Zhai B, Tan YX, Shan L, Liu Q, Chen HY, Dai RY, Qiu BJ, He YQ, Wang C, Zheng LY, Li YQ, Wu FQ, Li Z, Yan HX, Wang HY. Profound impact of gut homeostasis on chemically-induced pro-tumorigenic inflammation and hepatocarcinogenesis in rats. J Hepatol 2012; 57:803-12. [PMID: 22727732 DOI: 10.1016/j.jhep.2012.06.011] [Citation(s) in RCA: 214] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 06/06/2012] [Accepted: 06/06/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Due to its anatomic connection, the liver is constantly exposed to gut-derived bacterial products or metabolites. Disruption of gut homeostasis is associated with many human diseases. The aim of this study was to determine the role of gut homeostasis in initiation and progression of hepatocellular carcinoma (HCC). METHODS Disruption of intestinal homeostasis by penicillin or dextran sulfate sodium (DSS) and its restoration by probiotics were applied in a diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. RESULTS Patients with liver cirrhosis and HCC had significantly increased serum endotoxin levels. Chronic DEN treatment of rats was associated with an imbalance of subpopulations of the gut microflora including a significant suppression of Lactobacillus species, Bifidobacterium species and Enterococcus species as well as intestinal inflammation. Induction of enteric dysbacteriosis or intestinal inflammation by penicillin or DSS, respectively, significantly promoted tumor formation. Administration of probiotics dramatically mitigated enteric dysbacteriosis, ameliorated intestinal inflammation, and most importantly, decreased liver tumor growth and multiplicity. Interestingly, probiotics not only inhibited the translocation of endotoxin, which bears pathogen-associated molecular patterns (PAMPs) but also the activation of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1). As a result, the production of pro- and anti-inflammatory cytokines was skewed in favor of a reduced tumorigenic inflammation in the liver. CONCLUSIONS The data highlights the importance of gut homeostasis in the pathogenesis of HCC. Modulation of the gut microbiota by probiotics may represent a new avenue for therapeutic intervention to treat or prevent HCC development.
Collapse
Affiliation(s)
- Hui-Lu Zhang
- International Cooperation Laboratory on Signal Transduction, Liver Centre of SMMU, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wang F, Li Q, Wang C, Tang C, Li J. Dynamic alteration of the colonic microbiota in intestinal ischemia-reperfusion injury. PLoS One 2012; 7:e42027. [PMID: 22848694 PMCID: PMC3407053 DOI: 10.1371/journal.pone.0042027] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/29/2012] [Indexed: 12/15/2022] Open
Abstract
Background Intestinal ischemia-reperfusion (I/R) plays an important role in critical illnesses. Gut flora participate in the pathogenesis of the injury. This study is aimed at unraveling colonic microbiota alteration pattern and identifying specific bacterial species that differ significantly as well as observing colonic epithelium change in the same injury model during the reperfusion time course. Methodology/Principal Findings Denaturing gradient gel electrophoresis (DGGE) was used to monitor the colonic microbiota of control rats and experimental rats that underwent 0.5 hour ischemia and 1, 3, 6, 12, 24, and 72 hours following reperfusion respectively. The microbiota similarity, bacterial diversity and species that characterized the dysbiosis were estimated based on the DGGE profiles using a combination of statistical approaches. The interested bacterial species in the gel were cut and sequenced and were subsequently quantified and confirmed with real-time PCR. Meanwhile, the epithelial barrier was checked by microscopy and D-lactate analysis. Colonic flora changed early and differed significantly at 6 hours after reperfusion and then started to recover. The shifts were characterized by the increase of Escherichia coli and Prevotella oralis, and Lactobacilli proliferation together with epithelia healing. Conclusion/Significance This study shows for the first time that intestinal ischemia-reperfusion results in colonic flora dysbiosis that follows epithelia damage, and identifies the bacterial species that contribute most.
Collapse
Affiliation(s)
- Fan Wang
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chenyang Wang
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chun Tang
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- * E-mail:
| |
Collapse
|
42
|
Macia L, Thorburn AN, Binge LC, Marino E, Rogers KE, Maslowski KM, Vieira AT, Kranich J, Mackay CR. Microbial influences on epithelial integrity and immune function as a basis for inflammatory diseases. Immunol Rev 2012; 245:164-76. [PMID: 22168419 DOI: 10.1111/j.1600-065x.2011.01080.x] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Certain autoimmune diseases as well as asthma have increased in recent decades, particularly in developed countries. The hygiene hypothesis has been the prevailing model to account for this increase; however, epidemiology studies also support the contribution of diet and obesity to inflammatory diseases. Diet affects the composition of the gut microbiota, and recent studies have identified various molecules and mechanisms that connect diet, the gut microbiota, and immune responses. Herein, we discuss the effects of microbial metabolites, such as short chain fatty acids, on epithelial integrity as well as immune cell function. We propose that dysbiosis contributes to compromised epithelial integrity and disrupted immune tolerance. In addition, dietary molecules affect the function of immune cells directly, particularly through lipid G-protein coupled receptors such as GPR43.
Collapse
Affiliation(s)
- Laurence Macia
- Department of Immunology, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Snedeker SM, Hay AG. Do interactions between gut ecology and environmental chemicals contribute to obesity and diabetes? ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:332-9. [PMID: 22042266 PMCID: PMC3295356 DOI: 10.1289/ehp.1104204] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/31/2011] [Indexed: 05/17/2023]
Abstract
BACKGROUND Gut microbiota are important factors in obesity and diabetes, yet little is known about their role in the toxicodynamics of environmental chemicals, including those recently found to be obesogenic and diabetogenic. OBJECTIVES We integrated evidence that independently links gut ecology and environmental chemicals to obesity and diabetes, providing a framework for suggesting how these environmental factors may interact with these diseases, and identified future research needs. METHODS We examined studies with germ-free or antibiotic-treated laboratory animals, and human studies that evaluated how dietary influences and microbial changes affected obesity and diabetes. Strengths and weaknesses of studies evaluating how environmental chemical exposures may affect obesity and diabetes were summarized, and research gaps on how gut ecology may affect the disposition of environmental chemicals were identified. RESULTS Mounting evidence indicates that gut microbiota composition affects obesity and diabetes, as does exposure to environmental chemicals. The toxicology and pharmacology literature also suggests that interindividual variations in gut microbiota may affect chemical metabolism via direct activation of chemicals, depletion of metabolites needed for biotransformation, alteration of host biotransformation enzyme activities, changes in enterohepatic circulation, altered bioavailability of environmental chemicals and/or antioxidants from food, and alterations in gut motility and barrier function. CONCLUSIONS Variations in gut microbiota are likely to affect human toxicodynamics and increase individual exposure to obesogenic and diabetogenic chemicals. Combating the global obesity and diabetes epidemics requires a multifaceted approach that should include greater emphasis on understanding and controlling the impact of interindividual gut microbe variability on the disposition of environmental chemicals in humans.
Collapse
Affiliation(s)
- Suzanne M Snedeker
- Department of Microbiology and the Institute for Comparative and Environmental Toxicology, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
44
|
Martin R, Nauta AJ, Ben Amor K, Knippels LMJ, Knol J, Garssen J. Early life: gut microbiota and immune development in infancy. Benef Microbes 2011; 1:367-82. [PMID: 21831776 DOI: 10.3920/bm2010.0027] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The immune system of infants is actively downregulated during pregnancy and therefore the first months of life represent a period of heightened susceptibility to infection. After birth, there is an age-dependent maturation of the immune system. Exposure to environmental microbial components is suggested to play an important role in the maturation process. The gastrointestinal tract is the major site of interaction between the host immune system and microorganisms, both commensal as well as potentially pathogenic. It is well established that the mammalian immune system is designed to help protect the host from invading microorganisms and other danger signals. However, recent research is emerging in the field of host-microbe interactions showing that commensal microorganisms (microbiota) are most likely one of the drivers of immune development and, in turn the immune system shapes the composition of the microbiota. Specific early microbial exposure of the gut is thought to dramatically reduce the incidence of inflammatory, autoimmune and atopic diseases further fuelling the scientific view that microbial colonisation plays an important role in regulating and fine-tuning the immune system throughout life. Therefore, the use of pre-, pro- and synbiotics may result in a beneficial microbiota composition that might have a pivotal role on the prevention of several important diseases that develop in early life such as necrotizing enterocolitis and atopic eczema.
Collapse
Affiliation(s)
- R Martin
- Danone Research, Center for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Probiotic therapy: immunomodulating approach toward urinary tract infection. Curr Microbiol 2011; 63:484-90. [PMID: 21901556 DOI: 10.1007/s00284-011-0006-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/19/2011] [Indexed: 01/25/2023]
Abstract
Urinary tract infection (UTI) is an extremely common health problem, with an unpredictable history. Members of enterobacteriaceae family such as Escherichia coli, which are normal inhabitants of human intestines, account for the majority of these uncomplicated infections. Rarely, UTI can result from virus or fungus. There is a close correlation between loss of the normal genital microbiota, particularly Lactobacillus species, and an increased incidence of genital and bladder infections. Although antimicrobial agents are generally effective in eradicating these infections, there is a high incidence of recurrence. Use of Lactobacillus species to combat UTI is now giving modern concept of modern genitourinary vaccine with the facts that it not only maintains low pH of the genital area, produces hydrogen peroxide and hinders the growth of E. coli but also activates Toll-like receptor-2 (TLR2), which produces interleukin-10 (IL-10) and myeloid differentiation factor 88 (MyD88). E. coli activates TLR4, which is responsible for the activation of IL-12, extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). This process downregulates inflammatory reactions caused due to pathogens. Current review covers the probiotics-based TLR therapy and shed some knowledge for the use of Lactobacillus species as probiotics.
Collapse
|
46
|
Liu ZH, Shen TY, Zhang P, Ma YL, Moyer MP, Qin HL. Protective effects of Lactobacillus plantarum against epithelial barrier dysfunction of human colon cell line NCM460. World J Gastroenterol 2010; 16:5759-65. [PMID: 21128328 PMCID: PMC2997994 DOI: 10.3748/wjg.v16.i45.5759] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Lactobacillus plantarum (L. plantarum) in the intestinal permeability and expression of tight junction (TJ) using the normal human colon cell line NCM460.
METHODS: Paracellular permeability of NCM460 monolayers was determined by transepithelial electrical resistance and dextran permeability. Expression of TJ proteins in NCM460 cell monolayers was detected by Western blotting and quantitative real-time polymerase chain reaction.
RESULTS: L. plantarum played an important role in increasing transepithelial electrical resistance and decreasing the permeability to macromolecules of NCM460 monolayers against the disruption caused by enteropathogenic Escherichia coli (E. coli) or enteroinvasive E. coli. L. plantarum also prevented the decrease in the expression of TJ proteins and F-actin in NCM460 cells.
CONCLUSION: L. plantarum can protect against dysfunction of NCM460 intestinal epithelial barrier caused by enteropathogenic E. coli or enteroinvasive E. coli, and thus can be a potential candidate of therapeutic agents for the treatment of intestinal diseases.
Collapse
|
47
|
Lactobacillus plantarum surface layer adhesive protein protects intestinal epithelial cells against tight junction injury induced by enteropathogenic Escherichia coli. Mol Biol Rep 2010; 38:3471-80. [PMID: 21086172 DOI: 10.1007/s11033-010-0457-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Accepted: 11/09/2010] [Indexed: 01/01/2023]
Abstract
Lactobacillus plantarum (LP) has previously been used for the treatment and prevention of intestinal disorders and disease. However, the role of the LP surface layer adhesive protein (SLAP) in inhibition of epithelial cell disruption is not fully understood. The aim of the present study was to investigate the protective effects of purified SLAP on Caco-2 cells infected with enteropathogenic Escherichia coli (EPEC). The role of ERK in LP-mediated inhibition of tight junction (TJ) injury was also evaluated in order to determine the molecular mechanisms underlying the protective effects of LP in epithelial cells. SLAP was extracted and purified from LP cells using a porcine stomach mucin-Sepharose 4B column. SLAP-mediated inhibition of bacterial adhesion was measured using a competition-based adhesion assay. Expression of TJ-associated proteins, maintenance of TJ structure, and levels of extracellular signal regulated kinase (ERK) and ERK phosphorylation were assessed in SLAP-treated cells by a combination of real-time PCR, western blotting, and immunofluorescence microscopy. Cell permeability was analyzed by measurement of trans-epithelial electrical resistance (TER) and dextran permeability. The effect of SLAP on levels of apoptosis in epithelial cells was assessed by flow cytometry. Results from these experiments revealed that treatment with SLAP decreased the level of adhesion of EPEC to Caco-2 cells. SLAP treatment also enhanced expression of TJ proteins at both the mRNA and protein levels and affected F-actin distribution. Although ERK levels remained unchanged, ERK phosphorylation was increased by SLAP treatment. Caco-2 cells treated with SLAP exhibited increased TER and decreased macromolecular permeability, which was accompanied by a decrease in the level of apoptosis. Together, these results suggest that LP-produced SLAP protects intestinal epithelial cells from EPEC-induced injury, likely through a mechanism involving ERK activation.
Collapse
|
48
|
Arciero JC, Ermentrout GB, Upperman JS, Vodovotz Y, Rubin JE. Using a mathematical model to analyze the role of probiotics and inflammation in necrotizing enterocolitis. PLoS One 2010; 5:e10066. [PMID: 20419099 PMCID: PMC2856678 DOI: 10.1371/journal.pone.0010066] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 03/14/2010] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a severe disease of the gastrointestinal tract of pre-term babies and is thought to be related to the physiological immaturity of the intestine and altered levels of normal flora in the gut. Understanding the factors that contribute to the pathology of NEC may lead to the development of treatment strategies aimed at re-establishing the integrity of the epithelial wall and preventing the propagation of inflammation in NEC. Several studies have shown a reduced incidence and severity of NEC in neonates treated with probiotics (beneficial bacteria species). METHODOLOGY/PRINCIPAL FINDINGS The objective of this study is to use a mathematical model to predict the conditions under which probiotics may be successful in promoting the health of infants suffering from NEC. An ordinary differential equation model is developed that tracks the populations of pathogenic and probiotic bacteria in the intestinal lumen and in the blood/tissue region. The permeability of the intestinal epithelial layer is treated as a variable, and the role of the inflammatory response is included. The model predicts that in the presence of probiotics health is restored in many cases that would have been otherwise pathogenic. The timing of probiotic administration is also shown to determine whether or not health is restored. Finally, the model predicts that probiotics may be harmful to the NEC patient under very specific conditions, perhaps explaining the detrimental effects of probiotics observed in some clinical studies. CONCLUSIONS/SIGNIFICANCE The reduced, experimentally motivated mathematical model that we have developed suggests how a certain general set of characteristics of probiotics can lead to beneficial or detrimental outcomes for infants suffering from NEC, depending on the influences of probiotics on defined features of the inflammatory response.
Collapse
Affiliation(s)
- Julia C Arciero
- Department of Mathematics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America.
| | | | | | | | | |
Collapse
|
49
|
O'Flaherty S, Saulnier DM, Pot B, Versalovic J. How can probiotics and prebiotics impact mucosal immunity? Gut Microbes 2010; 1:293-300. [PMID: 21327037 PMCID: PMC3023613 DOI: 10.4161/gmic.1.5.12924] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 07/07/2010] [Indexed: 02/03/2023] Open
Abstract
The study of probiotics and prebiotics is an expanding field of interest and scientific research that has resulted in insights related to the host immune response. Recent advances have naturally led to key questions. What are the specific probiotic components that mediate immunomodulation? Can we extrapolate the results of in vitro studies in animal and human trials? Which biomarkers and immune parameters should be measured in probiotic and prebiotic intervention studies? These questions were part of a discussion entitled "How Can Probiotics and Prebiotics Impact Mucosal Immunity" at the 2009 Annual Meeting of the International Scientific Association for Probiotics and Prebiotics (ISAPP). This review highlights recent knowledge about the modulation of mucosal immunity by probiotics and prebiotics, as well as considerations for measuring their effects on mucosal immunity. A list of biomarkers and immune parameters to be measured in human clinical trials is included.
Collapse
Affiliation(s)
- Sarah O'Flaherty
- Department of Food, Bioprocessing and Nutrition Sciences; North Carolina State University; Raleigh, NC USA
| | - Delphine M Saulnier
- Department of Pathology and Immunology; Baylor College of Medicine; Texas Children's Hospital; Houston, TX USA,Department of Pathology and Texas Children's Microbiome Center; Texas Children's Hospital; Houston, TX USA
| | - Bruno Pot
- Bactéries Lactiques et Immunité des Muqueuses; Institut Pasteur de Lille; Lille, France
| | - James Versalovic
- Department of Pathology and Immunology; Baylor College of Medicine; Texas Children's Hospital; Houston, TX USA,Department of Pathology and Texas Children's Microbiome Center; Texas Children's Hospital; Houston, TX USA
| |
Collapse
|
50
|
Claud EC. Neonatal Necrotizing Enterocolitis -Inflammation and Intestinal Immaturity. Antiinflamm Antiallergy Agents Med Chem 2009; 8:248-259. [PMID: 20498729 PMCID: PMC2874244 DOI: 10.2174/187152309789152020] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Neonatal necrotizing enterocolitis is a devastating inflammatory bowel disease of premature infants. The pathogenesis remains incompletely understood and there is no specific treatment. Efforts are ongoing to understand aspects of intestinal immaturity which contribute to susceptibility to this disease. This review focuses on bacterial colonization patterns, intestinal barrier function, and inflammatory responses of immature enterocytes leading to a unique vulnerability of the preterm gut. In addition the possible therapeutic potential of factors in human milk and probiotic bacteria is discussed.
Collapse
Affiliation(s)
- Erika C. Claud
- The University of Chicago, Departments of Pediatrics and Medicine, Sections of Neonatology and Gastroenterology, 5841 S. Maryland Ave MC6060, Chicago, IL 60637, USA
| |
Collapse
|