101
|
Single Donor FMT Reverses Microbial/Immune Dysbiosis and Induces Clinical Remission in a Rat Model of Acute Colitis. Pathogens 2021; 10:pathogens10020152. [PMID: 33540919 PMCID: PMC7913212 DOI: 10.3390/pathogens10020152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Deviation in the gut microbial composition is involved in various pathologies, including inflammatory bowel disease (IBD). Faecal microbiota transplant (FMT) can act as a promising approach to treat IBD by which changes in microbiome can be reversed and homeostasis restored. Therefore, the aim of this study was to investigate the effect of FMT on the remission of acute inflammatory response using dextran sulfate sodium (DSS)-induced rat colitis model. Faecal microbial communities were analysed using the 16S rRNA approach, and clinical manifestations together with histological/haematological/biochemical/immunological analyses were assessed. Our study demonstrated significant shifts in the dominant species of microbiota under inflammatory conditions induced by DSS and evident restoration effect of FMT treatment on microbial composition. These faecal microbial alterations in FMT-treated rats led to a relative restoration of colon length, and a significant decrease in both epithelium damage and disease severity, which was reflected in lower serum pro-inflammatory cytokine levels. Haematological/biochemical parameters in DSS-treated animals showed signs of anaemia with a significant reduction in red blood cell count together with increasing levels of total bilirubin, creatinine and phosphorus suggesting potential protective effect of FMT. These results support FMT as a valuable therapeutic strategy to control inflammation during acute colitis.
Collapse
|
102
|
Gui DD, Luo W, Yan BJ, Ren Z, Tang ZH, Liu LS, Zhang JF, Jiang ZS. Effects of gut microbiota on atherosclerosis through hydrogen sulfide. Eur J Pharmacol 2021; 896:173916. [PMID: 33529724 DOI: 10.1016/j.ejphar.2021.173916] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/14/2021] [Accepted: 01/26/2021] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases are the leading cause of death and morbidity worldwide. Atherosclerotic cardiovascular disease (ASCVD) is affected by both environmental and genetic factors. Microenvironmental disorders of the human gut flora are associated with a variety of health problems, not only gastrointestinal diseases, such as inflammatory bowel disease, but also extralintestinal organs. Hydrogen sulfide (H2S) is the third gas signaling molecule other than nitric oxide and carbon monoxide. In the cardiovascular system, H2S plays important roles in the regulation of blood pressure, angiogenesis, smooth muscle cell proliferation and apoptosis, anti-oxidative stress, cardiac functions. This review is aiming to explore the potential role of gut microbiota in the development of atherosclerosis through hydrogen sulfide production as a novel therapeutic direction for atherosclerosis.
Collapse
Affiliation(s)
- Dan-Dan Gui
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Wen Luo
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Bin-Jie Yan
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Ji-Feng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China.
| |
Collapse
|
103
|
Citrus limon Peel Powder Reduces Intestinal Barrier Defects and Inflammation in a Colitic Murine Experimental Model. Foods 2021; 10:foods10020240. [PMID: 33503995 PMCID: PMC7912126 DOI: 10.3390/foods10020240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
This study examines the ameliorative effects of lemon (Citrus limon) peel (LP) powder on intestinal inflammation and barrier defects in dextran sulfate sodium (DSS)-induced colitic mice. The whole LP powder was fractionated into methanol (MetOH) extract and its extraction residue (MetOH residue), which were rich in polyphenolic compounds and dietary fibers, respectively. Mice were fed diets containing whole LP powder, MetOH extract, and MetOH residue for 16 d. DSS administration for 9 d induced bodyweight loss, reduced colon length, reduced the colonic expression of tight junction proteins including zonula occludens-1 and -2, and claudin-3 and -7, and upregulated colonic mRNA expression of interleukin 6, chemokine (C-X-C motif) ligand 2, and C-C motif chemokine ligand 2. Feeding LP powder restored these abnormalities, and the MetOH residue, but not MetOH extract, also showed similar restorations. Feeding LP powder and MetOH residue increased fecal concentrations of acetate and n-butyrate. Taken together, LP powder reduced intestinal damage through the protection of tight junction barriers and suppressed an inflammatory reaction in colitic mice. These results suggest that acetate and n-butyrate produced from the microbial metabolism of dietary fibers in LP powder contributed to reducing colitis.
Collapse
|
104
|
Microbiota, a New Playground for the Omega-3 Polyunsaturated Fatty Acids in Cardiovascular Diseases. Mar Drugs 2021; 19:md19020054. [PMID: 33498729 PMCID: PMC7931107 DOI: 10.3390/md19020054] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Several cardioprotective mechanisms attributed to Omega-3 polyunsaturated fatty acids (PUFAs) have been studied and widely documented. However, in recent years, studies have supported the concept that the intestinal microbiota can play a much larger role than we had anticipated. Microbiota could contribute to several pathologies, including cardiovascular diseases. Indeed, an imbalance in the microbiota has often been reported in patients with cardiovascular disease and produces low-level inflammation. This inflammation contributes to, more or less, long-term development of cardiovascular diseases. It can also worsen the symptoms and the consequences of these pathologies. According to some studies, omega-3 PUFAs in the diet could restore this imbalance and mitigate its harmful effects on cardiovascular diseases. Many mechanisms are involved and included: (1) a reduction of bacteria producing trimethylamine (TMA); (2) an increase in bacteria producing butyrate, which has anti-inflammatory properties; and (3) a decrease in the production of pro-inflammatory cytokines. Additionally, omega-3 PUFAs would help maintain better integrity in the intestinal barrier, thereby preventing the translocation of intestinal contents into circulation. This review will summarize the effects of omega-3 PUFAs on gut micro-biota and the potential impact on cardiac health.
Collapse
|
105
|
Zhang J, Wan J, Chen D, Yu B, He J. Low-Molecular-Weight Chitosan Attenuates Lipopolysaccharide-Induced Inflammation in IPEC-J2 Cells by Inhibiting the Nuclear Factor-κB Signalling Pathway. Molecules 2021; 26:molecules26030569. [PMID: 33499133 PMCID: PMC7865926 DOI: 10.3390/molecules26030569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023] Open
Abstract
Low-molecular-weight chitosan (LMWC), a product of chitosan deacetylation, possesses anti-inflammatory effects. In the present study, a porcine small intestinal epithelial cell line, IPEC-J2, was used to assess the protective effects of LMWC on lipopolysaccharide (LPS)-induced intestinal epithelial cell injury. IPEC-J2 cells were pretreated with or without LMWC (400 μg/mL) in the presence or absence of LPS (5 μg/mL) for 6 h. LMWC pretreatment increased (p < 0.05) the occludin abundance and decreased (p < 0.05) the tumour necrosis factor-α (TNF-α) production, apoptosis rate and cleaved cysteinyl aspartate-specific protease-3 (caspase-3) and -8 contents in LPS-treated IPEC-J2 cells. Moreover, LMWC pretreatment downregulated (p < 0.05) the expression levels of TNF receptor 1 (TNFR1) and TNFR-associated death domain and decreased (p < 0.05) the nuclear and cytoplasmic abundance of nuclear factor-κB (NF-κB) p65 in LPS-stimulated IPEC-J2 cells. These results suggest that LMWC exerts a mitigation effect on LPS-induced intestinal epithelial cell damage by suppressing TNFR1-mediated apoptosis and decreasing the production of proinflammatory cytokines via the inhibition of NF-κB signalling pathway.
Collapse
Affiliation(s)
| | | | | | | | - Jun He
- Correspondence: ; Tel.: +86-13419354223; Fax: +86-28-86291781
| |
Collapse
|
106
|
Barroso FAL, de Jesus LCL, de Castro CP, Batista VL, Ferreira Ê, Fernandes RS, de Barros ALB, Leclerq SY, Azevedo V, Mancha-Agresti P, Drumond MM. Intake of Lactobacillus delbrueckii (pExu: hsp65) Prevents the Inflammation and the Disorganization of the Intestinal Mucosa in a Mouse Model of Mucositis. Microorganisms 2021; 9:microorganisms9010107. [PMID: 33466324 PMCID: PMC7824804 DOI: 10.3390/microorganisms9010107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
5-Fluorouracil (5-FU) is an antineoplastic drug that causes, as a side effect, intestinal mucositis, acute inflammation in the small bowel. The Heat Shock Protein (Hsp) are highly expressed in inflammatory conditions, developing an important role in immune modulation. Thus, they are potential candidates for the treatment of inflammatory diseases. In the mucositis mouse model, the present study aimed to evaluate the beneficial effect of oral administration of milk fermented by Lactobacillus delbrueckii CIDCA 133 (pExu:hsp65), a recombinant strain. This approach showed increased levels of sIgA in the intestinal fluid, reducing inflammatory infiltrate and intestinal permeability. Additionally, the histological score was improved. Protection was associated with a reduction in the gene expression of pro-inflammatory cytokines such as Tnf, Il6, Il12, and Il1b, and an increase in Il10, Muc2, and claudin 1 (Cldn1) and 2 (Cldn2) gene expression in ileum tissue. These findings are corroborated with the increased number of goblet cells, the electronic microscopy images, and the reduction of intestinal permeability. The administration of milk fermented by this recombinant probiotic strain was also able to reverse the high levels of gene expression of Tlrs caused by the 5-FU. Thus, the rCIDCA 133:Hsp65 strain was revealed to be a promising preventive strategy for small bowel inflammation.
Collapse
Affiliation(s)
- Fernanda Alvarenga Lima Barroso
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Camila Prosperi de Castro
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Viviane Lima Batista
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Ênio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Renata Salgado Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - André Luís Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - Sophie Yvette Leclerq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel Dias (FUNED), Belo Horizonte 30510-010, Brazil;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Faculdade de Minas-Faminas-BH, Medicina, Belo Horizonte 31744-007, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| | - Mariana Martins Drumond
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte 31421-169, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| |
Collapse
|
107
|
Rea K, O' Mahony SM, Cryan JF. High and Mighty? Cannabinoids and the microbiome in pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 9:100061. [PMID: 33665479 PMCID: PMC7905370 DOI: 10.1016/j.ynpai.2021.100061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/21/2020] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
In this review, we will focus on the potential role of the endogenous cannabinoids in modulating microbiota-driven changes in peripheral and central pain processing. We also focus on the overlap in mechanisms whereby commensal gut microbiota and endocannabinoid ligands can regulate inflammation and further aim to exploit our understanding of their role in microbiota-gut-brain axis communication in pain processing.
Within the human gut, we each harbour a unique ecosystem represented by trillions of microbes that contribute to our health and wellbeing. These gut microbiota form part of a complex network termed the microbiota-gut-brain axis along with the enteric nervous system, sympathetic and parasympathetic divisions of the autonomic nervous system, and neuroendocrine and neuroimmune components of the central nervous system. Through endocrine, immune and neuropeptide/neurotransmitter systems, the microbiota can relay information about health status of the gut. This in turn can profoundly impact neuronal signalling not only in the periphery, but also in the brain itself and thus impact on emotional systems and behavioural responses. This may be true for pain, as the top-down facilitation or inhibition of pain processing occurs at a central level, while ascending afferent nociceptive information from the viscera and systemic areas travel through the periphery and spinal cord to the brain. The endogenous cannabinoid receptors are ubiquitously expressed throughout the gut, periphery and in brain regions associated with pain responding, and represent targets for endogenous and exogenous manipulation. In this review, we will focus on the potential role of the endogenous cannabinoids in modulating microbiota-driven changes in peripheral and central pain processing. We also focus on the overlap in mechanisms whereby commensal gut microbiota and endocannabinoid ligands can regulate inflammation and further aim to exploit our understanding of their role in microbiota-gut-brain axis communication in pain processing.
Collapse
Affiliation(s)
- Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Siobhain M O' Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
108
|
Tian M, Chen J, Wu Z, Song H, Yang F, Cui C, Chen F, Zhang S, Guan W. Fat Encapsulation Reduces Diarrhea in Piglets Partially by Repairing the Intestinal Barrier and Improving Fatty Acid Transport. Animals (Basel) 2020; 11:ani11010028. [PMID: 33375218 PMCID: PMC7824132 DOI: 10.3390/ani11010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
(1) Background: Nutritional strategies to enhance gut function and reduce the piglet diarrhea rate are critical to increase the growth performance of piglets. The purpose of this study was to investigate whether dietary fat types and/or fat microencapsulation techniques are involved in regulating the fatty acid transport system and the mechanical and immunological barriers of the small intestine. (2) Methods: Three hundred twenty-four weaning piglets were randomly divided into three groups fed a soybean oil diet (SBO, control group, 6.0% soybean oil), palm oil diet (PO, 6.0% palm oil), or encapsulated palm oil diet (EPO, 7.5% encapsulated palm oil). (3) Results: A significantly lower mRNA expression of the claudin was observed in the duodenum and jejunum of the PO group than in the SBO group (p < 0.05). However, the mRNA expression and protein abundance of claudin and ZO-1 in the jejunum of the EPO group were higher (p < 0.05) than in the PO group. Porcine β-defensin (pBD) secretion was not significantly different between the SBO and PO groups (p > 0.05), while the pBD-2 levels were significantly different (p < 0.05). Compared with the PO group, the EPO group exhibited a significantly increased secretion of pBD-2 and pBD-129 in the small intestine (p < 0.05) and pBD-1 in the jejunum and ileum (p < 0.05). The protein abundances of apolipoprotein AIV (Apo AIV) and intestinal fatty acid binding protein (I-FABP) were significantly lower in the PO group than in the SBO group (p < 0.05). Simultaneously, the protein abundances of fatty acid transport protein 4 (FATP4), fatty acid translocase (CD36), and I-FABP were higher in the EPO group than in the PO group. Furthermore, the low digestibility of palm oil (PO group) might negatively regulate intestinal tight junctions, fatty acid transporters, lipoproteins, and β-defensin through the activation of the AMPK/mTORC1 and AMPK/Sirt1/NF-κB pathways. (4) Conclusions: In summary, microencapsulation techniques might alleviate the negative effects of palm oil and help to improve the intestinal fatty acid transport system and barrier function.
Collapse
Affiliation(s)
- Min Tian
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Jiaming Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Zhihui Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Hanqing Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Fei Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Chang Cui
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
| | - Fang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (S.Z.); (W.G.); Tel./Fax: +86-20-85284837 (S.Z.); +86-20-85284837 (W.G.)
| | - Wutai Guan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (M.T.); (J.C.); (Z.W.); (H.S.); (F.Y.); (C.C.); (F.C.)
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (S.Z.); (W.G.); Tel./Fax: +86-20-85284837 (S.Z.); +86-20-85284837 (W.G.)
| |
Collapse
|
109
|
Zhang Y, Wang Z, Peng J, Gerner ST, Yin S, Jiang Y. Gut microbiota-brain interaction: An emerging immunotherapy for traumatic brain injury. Exp Neurol 2020; 337:113585. [PMID: 33370556 DOI: 10.1016/j.expneurol.2020.113585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023]
Abstract
Individuals suffering from traumatic brain injury (TBI) often experience the activation of the immune system, resulting in declines in cognitive and neurological function after brain injury. Despite decades of efforts, approaches for clinically effective treatment are sparse. Evidence on the association between current therapeutic strategies and clinical outcomes after TBI is limited to poorly understood mechanisms. For decades, an increasing number of studies suggest that the gut-brain axis (GBA), a bidirectional communication system between the central nervous system (CNS) and the gastrointestinal tract, plays a critical role in systemic immune response following neurological diseases. In this review, we detail current knowledge of the immune pathologies of GBA after TBI. These processes may provide a new therapeutic target and rehabilitation strategy developed and used in clinical treatment of TBI patients.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhaoyang Wang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Stefan T Gerner
- Department of Neurology, University Hospital Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
110
|
Lv Q, Xu D, Zhang X, Yang X, Zhao P, Cui X, Liu X, Yang W, Yang G, Xing S. Association of Hyperuricemia With Immune Disorders and Intestinal Barrier Dysfunction. Front Physiol 2020; 11:524236. [PMID: 33329010 PMCID: PMC7729003 DOI: 10.3389/fphys.2020.524236] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background More than 30–40% of uric acid is excreted via the intestine, and the dysfunction of intestinal epithelium disrupts uric acid excretion. The involvement of gut microbiota in hyperuricemia has been reported in previous studies, but the changes and mechanisms of intestinal immunity in hyperuricemia are still unknown. Methods This study developed a urate oxidase (Uox)-knockout (Uox–/–) mouse model for hyperuricemia using CRISPR/Cas9 technology. The lipometabolism was assessed by measuring changes in biochemical indicators. Furthermore, 4-kDa fluorescein isothiocyanate–labeled dextran was used to assess gut barrier function. Also, 16S rRNA sequencing was performed to examine the changes in gut microbiota in mouse feces. RNA sequencing, Western blot, Q-PCR, ELISA, and immunohistochemical analysis were used for measuring gene transcription, the number of immune cells, and the levels of cytokines in intestinal tissues, serum, kidney, liver, pancreas, and vascellum. Results This study showed that the abundance of inflammation-related microbiota increased in hyperuricemic mice. The microbial pattern recognition–associated Toll-like receptor pathway and inflammation-associated TNF and NF-kappa B signaling pathways were significantly enriched. The increased abundance of inflammation-related microbiota resulted in immune disorders and intestinal barrier dysfunction by upregulating TLR2/4/5 and promoting the release of IL-1β and TNF-α. The levels of epithelial tight junction proteins occludin and claudin-1 decreased. The expression of the pro-apoptotic gene Bax increased. The levels of LPS and TNF-α in systemic circulation increased in hyperuricemic mice. A positive correlation was observed between the increase in intestinal permeability and serum levels of uric acid. Conclusion Hyperuricemia was characterized by dysregulated intestinal immunity, compromised intestinal barrier, and systemic inflammation. These findings might serve as a basis for future novel therapeutic interventions for hyperuricemia.
Collapse
Affiliation(s)
- Qiulan Lv
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Daxing Xu
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuezhi Zhang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaomin Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Zhao
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuena Cui
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiu Liu
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wan Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guanpin Yang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Shichao Xing
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, China.,Institute of Sports Medicine and Health, Qingdao University, Qingdao, China.,Guy's & St Thomas' Hospital, King's College London, London, United Kingdom
| |
Collapse
|
111
|
Shi J, Du P, Xie Q, Wang N, Li H, Smith EE, Li C, Liu F, Huo G, Li B. Protective effects of tryptophan-catabolizing Lactobacillus plantarum KLDS 1.0386 against dextran sodium sulfate-induced colitis in mice. Food Funct 2020; 11:10736-10747. [PMID: 33231244 DOI: 10.1039/d0fo02622k] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tryptophan is an essential amino acid for the human body, whose intake is through the diet. Several studies support the theory that microbiota-derived tryptophan metabolite played a crucial role in maintaining the balance between gut microbiota and the mucosal immune system. Previously, we selected the Lactobacillus plantarum KLDS 1.0386 strain with high tryptophan-metabolic activity after the screening of 16 Lactobacillus strains. The current study aimed to assess the effects of L. plantarum KLDS 1.0386 combination with tryptophan in improving ulcerative colitis (UC) induced by dextran sodium sulfate (DSS) and the potential mechanisms involved. Our results showed that L. plantarum KLDS 1.0386 combined with tryptophan (LAB + Trp) decreased DAI score, MPO level, and pro-inflammatory cytokine (TNF-α, IL-1β, and IL-6) concentration. It also increased anti-inflammatory cytokine (IL-10) production, tight junction proteins (claudin-1, occludin, and ZO-1), and mucin (MUC1 and MUC2) mRNA expressions. The level of indole-3-acetic acid (IAA), an important tryptophan metabolite in the liver, serum, and colon, was elevated after LAB + Trp treatment, which further upregulated aryl hydrocarbon receptor (AHR) mRNA expression to activate the IL-22/STAT3 signaling pathway. Moreover, the supplementation with LAB + Trp modulated gut microbiota composition. The present study provided novel insights that can be used to reduce the number of UC patients by employing a method utilizing tryptophan-catabolizing Lactobacillus strains.
Collapse
Affiliation(s)
- Jialu Shi
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Ma T, Peng W, Liu Z, Gao T, Liu W, Zhou D, Yang K, Guo R, Duan Z, Liang W, Bei W, Yuan F, Tian Y. Tea polyphenols inhibit the growth and virulence of ETEC K88. Microb Pathog 2020; 152:104640. [PMID: 33232763 DOI: 10.1016/j.micpath.2020.104640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 01/20/2023]
Abstract
Diarrhea caused by Enterotoxigenic Escherichia coli (ETEC) causes high levels of morbidity and mortality in neonatal piglets. Owing to the abuse of antibiotics and emergence of drug resistance, antibiotics are no longer considered only beneficial, but also potentially harmful drugs. Supplements that can inhibit the growth of bacteria are expected to replace antibiotics. Tea polyphenols have numerous important biological functions, including antibacterial, antiviral, antioxidative, anti-inflammatory, and antihypertensive effects. We investigated the role of tea polyphenols in ETEC K88 infection using a mouse model. Pretreating with tea polyphenols attenuated the symptoms induced by ETEC K88. Furthermore, in a cell adherence assay, tea polyphenols inhibited ETEC K88 adherence to IPEC-J2 cells. When cells were infected with ETEC K88, mRNA and protein levels of claudin-1 were significantly decreased compared with those of control cells. However, when cells were pretreated with tea polyphenols, claudin-1 mRNA and protein levels were higher than those in cells without pretreatment upon cell infection with ETEC K88. TLR2 mRNA levels were also higher following cell infection with ETEC K88 when cells were pretreated with tea polyphenols. These data revealed that tea polyphenols could increase the barrier integrity of IPEC-J2 cells by upregulating expression of claudin-1 through activation of TLR2. Tea polyphenols had beneficial effects on epithelial barrier function. Therefore, tea polyphenols could be used as a novel strategy to control and treat pig infections caused by ETEC K88.
Collapse
Affiliation(s)
- Tianfeng Ma
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Cooperative Innovation Center of Sustainable Pig Production, Wuhan, 430070, China
| | - Wei Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Cooperative Innovation Center of Sustainable Pig Production, Wuhan, 430070, China
| | - Zewen Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Ting Gao
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Wei Liu
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Danna Zhou
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Keli Yang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Rui Guo
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Zhengying Duan
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Wan Liang
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China
| | - Weicheng Bei
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Cooperative Innovation Center of Sustainable Pig Production, Wuhan, 430070, China.
| | - Fangyan Yuan
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China.
| | - Yongxiang Tian
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture and Rural Affairs), Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China; Hubei Provincial Key Laboratory of Animal Pathogenic Microbiology, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan, 430064, China.
| |
Collapse
|
113
|
Metabolomics analysis of microbiota-gut-brain axis in neurodegenerative and psychiatric diseases. J Pharm Biomed Anal 2020; 194:113681. [PMID: 33279302 DOI: 10.1016/j.jpba.2020.113681] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/30/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Gut microbiota represents a complex physiological ecosystem that influences the host health. Alterations in the microbiome metabolism affect the body homeostasis and they have been associated with the development of different human neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease, autism spectrum disorder, bipolar disorder, depression, Huntington's disease, Parkinson's disease, posttraumatic stress disorder and schizophrenia. The development of these complex diseases is influenced by various factors, including genetic predisposition and environmental triggers. Gut microbiota has recently emerged as an important actor in their physiopathology that has been shown to play a role in inflammation, oxidative stress, and gut permeability. Therefore, targeting the metabolites that are produced by or associated with the gut microbiota may help us understand how imbalance in the gut-brain axis affects human health. This review offers a comprehensive overview of the literature on this matter, offering the readers an insight in the state-of-art metabolic measurements of the gut-brain axis in various brain-related diseases.
Collapse
|
114
|
Hai Y, Zhang Y, Liang Y, Ma X, Qi X, Xiao J, Xue W, Luo Y, Yue T. Advance on the absorption, metabolism, and efficacy exertion of quercetin and its important derivatives. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.50] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yu Hai
- College of Food Science and Technology Northwest University Xi'an Shaanxi P. R. China
| | - Yuanxiao Zhang
- School of Chemical Engineering Northwest University Xi'an Shaanxi P. R. China
| | - Yingzhi Liang
- College of Food Science and Technology Northwest University Xi'an Shaanxi P. R. China
| | - Xiaoyu Ma
- College of Life Science Northwest University Xi'an Shaanxi P. R. China
| | - Xiao Qi
- College of Food Science and Technology Northwest University Xi'an Shaanxi P. R. China
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology University of Vigo ‐ Ourense Campus Ourense E‐32004 Spain
| | - Weiming Xue
- School of Chemical Engineering Northwest University Xi'an Shaanxi P. R. China
| | - Yane Luo
- College of Food Science and Technology Northwest University Xi'an Shaanxi P. R. China
| | - Tianli Yue
- College of Food Science and Technology Northwest University Xi'an Shaanxi P. R. China
- Laboratory of Quality and Safety Risk Assessment for Agro‐products (Yangling) Ministry of Agriculture Beijing P. R. China
| |
Collapse
|
115
|
Kim SM, Song IH. The clinical impact of gut microbiota in chronic kidney disease. Korean J Intern Med 2020; 35:1305-1316. [PMID: 32872729 PMCID: PMC7652652 DOI: 10.3904/kjim.2020.411] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Gut microorganisms play critical roles in both maintaining host homeostasis and the development of diverse diseases. Gut dysbiosis, an alteration of the composition and function of gut microorganisms, is commonly seen in patients with chronic kidney disease (CKD). CKD itself contributes to a disruption of the symbiotic relationship between the gut microbiota and the host, while the resulting gut dysbiosis may play a part in stage progression of CKD. This bidirectional relationship supports the concept that the gut microbiota is considered a novel focus for the pathogenesis and management of CKD. This article examines the interaction between the gut microbiota and the kidney, the mutual effects of dysbiosis and CKD, and possible treatment options to restore gut eubiosis, and reduce CKD progression and its related complications.
Collapse
Affiliation(s)
- So Mi Kim
- Division of Nephrology, Department of Internal Medicine, Dankook University Hospital, Cheonan, Korea
| | - Il han Song
- Division of Hepatology, Department of Internal Medicine, Dankook University Hospital, Cheonan, Korea
- Correspondence to Il Han Song, M.D. Division of Hepatology, Department of Internal Medicine, Dankook University Hospital, 201 Manghyang-ro, Dongnam-gu, Cheonan 31116, Korea Tel: +82-41-550-3924 Fax: +82-41-556-3256 E-mail:
| |
Collapse
|
116
|
Yuan D, Wang J, Xiao D, Li J, Liu Y, Tan B, Yin Y. Eucommia ulmoides Flavones as Potential Alternatives to Antibiotic Growth Promoters in a Low-Protein Diet Improve Growth Performance and Intestinal Health in Weaning Piglets. Animals (Basel) 2020; 10:E1998. [PMID: 33143126 PMCID: PMC7694009 DOI: 10.3390/ani10111998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023] Open
Abstract
Eucommia ulmoides flavones (EUF) have been demonstrated to attenuate the inflammation and oxidative stress of piglets. This study aimed to test whether EUF could be used as an alternative antibiotic growth promoter to support growth performance and maintain intestinal health in weanling piglets. Weaned piglets (n = 480) were assigned into three groups and fed with a low-protein basal diet (NC), or supplementation with antibiotics (PC) or 0.01% EUF (EUF). Blood, intestinal contents, and intestine were collected on days 15 and 35 after weaning. The results showed the PC and EUF supplementations increased (p < 0.05) body weight on day 35, average daily gain and gain: feed ratio from day 15 to day 35 and day 0 to day 35, whereas decreased (p < 0.05) the diarrhea index of weanling piglets. EUF treatment increased (p < 0.05) jejunal villus height: crypt depth ratio, jejunal and ileal villus height, and population of ileal lactic acid bacteria on day 15 but decreased (p < 0.05) the population of ileal coliform bacteria on day 15 and day 35. These findings indicated the EUF, as the potential alternative to in-feed antibiotic growth promoter, could improve growth performance and intestinal morphology, and decrease colonization of coliform bacteria and diarrhea index in weanling piglets.
Collapse
Affiliation(s)
- Daixiu Yuan
- Department of Medicine, Jishou University, Jishou 416000, China;
| | - Jing Wang
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Dingfu Xiao
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
| | - Jiefeng Li
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA;
| | - Bie Tan
- Department of Animal Science, Hunan Agricultural University, Changsha 410000, China; (D.X.); (B.T.)
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| |
Collapse
|
117
|
Zhang H, Liu M, Liu X, Zhong W, Li Y, Ran Y, Guo L, Chen X, Zhao J, Wang B, Zhou L. Bifidobacterium animalis ssp. Lactis 420 Mitigates Autoimmune Hepatitis Through Regulating Intestinal Barrier and Liver Immune Cells. Front Immunol 2020; 11:569104. [PMID: 33123141 PMCID: PMC7573389 DOI: 10.3389/fimmu.2020.569104] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated inflammatory liver disease of uncertain cause. Accumulating evidence shows that gut microbiota and intestinal barrier play significant roles in AIH thus the gut–liver axis has important clinical significance as a potential therapeutic target. In the present study, we found that Bifidobacterium animalis ssp. lactis 420 (B420) significantly alleviated S100-induced experimental autoimmune hepatitis (EAH) and modulated the gut microbiota composition. While the analysis of clinical specimens revealed that the fecal SCFA quantities were decreased in AIH patients, and B420 increased the cecal SCFA quantities in EAH mice. Remarkably, B420 application improved intestinal barrier function through upregulation of tight junction proteins in both vitro and vivo experiments. Moreover, B420 decreased the serum endotoxin level and suppressed the RIP3 signaling pathway of liver macrophages in EAH mice thus regulated the proliferation of Th17 cells. Nevertheless, the inhibition effect of B420 on RIP3 signaling pathway was blunted in vitro studies. Together, our results showed that early intervention with B420 contributed to improve the liver immune homeostasis and liver injury in EAH mice, which might be partly due to the protection of intestinal barrier. Our study suggested the potential efficacy of probiotics application against AIH and the promising therapeutic strategies targeting gut–liver axis for AIH.
Collapse
Affiliation(s)
- Hongxia Zhang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Man Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xin Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yanni Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ying Ran
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Liping Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xu Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Department of Gastroenterology and Hepatology, People's Hospital of Hetian District, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
118
|
Yao L, Tang Y, Chen B, Hong W, Xu X, Liu Y, Aguilar ZP, Xu H. Oral exposure of titanium oxide nanoparticles induce ileum physical barrier dysfunction via Th1/Th2 imbalance. ENVIRONMENTAL TOXICOLOGY 2020; 35:982-990. [PMID: 32333507 DOI: 10.1002/tox.22934] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/09/2020] [Accepted: 04/04/2020] [Indexed: 05/28/2023]
Abstract
In this work, we aimed to evaluate the adverse effects and the mechanism of intestinal barrier caused by titanium dioxide nanoparticles (TiO2 NPs). Here, the effects of two different dosages (300 and 1200 mg/kg) of TiO2 NPs on female mice (n = 5) were investigated. After 28-day oral exposure, the results of Ti content were significantly increased in the ileum in comparison with the control. The histopathological structure index of the ileum was significantly changed after TiO2 NPs exposure; villi height and crypt depth were decreased and increased, respectively. Meanwhile, TiO2 NPs treatment also significantly altered the transcription levels of genes. First, the GATA-3 and STAT-4 were upregulation and downregulation, respectively. Second, gene expressions of the Zonula Occludens-1, claudin (CLDN)-12, occludin, and myosin light chain kinase were significantly upregulated, while the CLDN-3 was decreased. Finally, the caspase-3, caspase-9, and caspase-12 were upregulated. The results of TUNEL staining indicated apoptosis in the ileum. In general, TiO2 NPs treatment significantly changed the intestine physical barrier in a dose-dependent manner. The toxicity of TiO2 NPs could be through the imbalance in the Th1/Th2.
Collapse
Affiliation(s)
- Liyang Yao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yizhou Tang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Bolu Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Wuding Hong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xinyue Xu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Liu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | | | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
119
|
Heart Failure Disturbs Gut-Blood Barrier and Increases Plasma Trimethylamine, a Toxic Bacterial Metabolite. Int J Mol Sci 2020; 21:ijms21176161. [PMID: 32859047 PMCID: PMC7504565 DOI: 10.3390/ijms21176161] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Trimethylamine (TMA) is a gut bacteria product oxidized by the liver to trimethylamine-N-oxide (TMAO). Clinical evidence suggests that cardiovascular disease is associated with increased plasma TMAO. However, little headway has been made in understanding this relationship on a mechanistic and molecular level. We investigated the mechanisms affecting plasma levels of TMAO in Spontaneously Hypertensive Heart Failure (SHHF) rats. Healthy Wistar Kyoto (WKY) and SHHF rats underwent metabolic, hemodynamic, histopathological and biochemical measurements, including tight junction proteins analysis. Stool, plasma and urine samples were evaluated for TMA and TMAO using ultra performance liquid chromatography-mass spectrometry. SHHF presented disturbances of the gut–blood barrier including reduced intestinal blood flow, decreased thickness of the colonic mucosa and alterations in tight junctions, such as claudin 1 and 3, and zonula occludens-1. This was associated with significantly higher plasma levels of TMA and TMAO and increased gut-to-blood penetration of TMA in SHHF compared to WKY. There was no difference in kidney function or liver oxidation of TMA to TMAO between WKY and SHHF. In conclusion, increased plasma TMAO in heart failure rats results from a perturbed gut–blood barrier and increased gut-to-blood passage of TMAO precursor, i.e., TMA. Increased gut-to-blood penetration of bacterial metabolites may be a marker and a mediator of cardiovascular pathology.
Collapse
|
120
|
Zhang J, Huang YJ, Yoon JY, Kemmitt J, Wright C, Schneider K, Sphabmixay P, Hernandez-Gordillo V, Holcomb SJ, Bhushan B, Rohatgi G, Benton K, Carpenter D, Kester JC, Eng G, Breault DT, Yilmaz O, Taketani M, Voigt CA, Carrier RL, Trumper DL, Griffith LG. Primary human colonic mucosal barrier crosstalk with super oxygen-sensitive Faecalibacterium prausnitzii in continuous culture. MED 2020; 2:74-98.e9. [PMID: 33511375 DOI: 10.1016/j.medj.2020.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background The gut microbiome plays an important role in human health and disease. Gnotobiotic animal and in vitro cell-based models provide some informative insights into mechanistic crosstalk. However, there is no existing system for a long-term co-culture of a human colonic mucosal barrier with super oxygen-sensitive commensal microbes, hindering the study of human-microbe interactions in a controlled manner. Methods Here, we investigated the effects of an abundant super oxygen-sensitive commensal anaerobe, Faecalibacterium prausnitzii, on a primary human mucosal barrier using a Gut-MIcrobiome (GuMI) physiome platform that we designed and fabricated. Findings Long-term continuous co-culture of F. prausnitzii for two days with colon epithelia, enabled by continuous flow of completely anoxic apical media and aerobic basal media, resulted in a strictly anaerobic apical environment fostering growth of and butyrate production by F. prausnitzii, while maintaining a stable colon epithelial barrier. We identified elevated differentiation and hypoxia-responsive genes and pathways in the platform compared with conventional aerobic static culture of the colon epithelia, attributable to a combination of anaerobic environment and continuous medium replenishment. Furthermore, we demonstrated anti-inflammatory effects of F. prausnitzii through HDAC and the TLR-NFKB axis. Finally, we identified that butyrate largely contributes to the anti-inflammatory effects by downregulating TLR3 and TLR4. Conclusions Our results are consistent with some clinical observations regarding F. prausnitzii, thus motivating further studies employing this platform with more complex engineered colon tissues for understanding the interaction between the human colonic mucosal barrier and microbiota, pathogens, or engineered bacteria.
Collapse
Affiliation(s)
| | | | - Jun Young Yoon
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,School of Mechanical Engineering, Yonsei University, Seoul 03722, South Korea
| | | | | | | | | | | | | | - Brij Bhushan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gar Rohatgi
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | - Kyle Benton
- EPAM Continuum, 41 University Drive, Newtown, PA 18940, USA
| | | | | | | | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Rebecca L Carrier
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - David L Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G Griffith
- Department of Biological Engineering.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
121
|
Kappler K, Hennet T. Emergence and significance of carbohydrate-specific antibodies. Genes Immun 2020; 21:224-239. [PMID: 32753697 PMCID: PMC7449879 DOI: 10.1038/s41435-020-0105-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Carbohydrate-specific antibodies are widespread among all classes of immunoglobulins. Despite their broad occurrence, little is known about their formation and biological significance. Carbohydrate-specific antibodies are often classified as natural antibodies under the assumption that they arise without prior exposure to exogenous antigens. On the other hand, various carbohydrate-specific antibodies, including antibodies to ABO blood group antigens, emerge after the contact of immune cells with the intestinal microbiota, which expresses a vast diversity of carbohydrate antigens. Here we explore the development of carbohydrate-specific antibodies in humans, addressing the definition of natural antibodies and the production of carbohydrate-specific antibodies upon antigen stimulation. We focus on the significance of the intestinal microbiota in shaping carbohydrate-specific antibodies not just in the gut, but also in the blood circulation. The structural similarity between bacterial carbohydrate antigens and surface glycoconjugates of protists, fungi and animals leads to the production of carbohydrate-specific antibodies protective against a broad range of pathogens. Mimicry between bacterial and human glycoconjugates, however, can also lead to the generation of carbohydrate-specific antibodies that cross-react with human antigens, thereby contributing to the development of autoimmune disorders.
Collapse
Affiliation(s)
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
122
|
Forkosh E, Kenig A, Ilan Y. Introducing variability in targeting the microtubules: Review of current mechanisms and future directions in colchicine therapy. Pharmacol Res Perspect 2020; 8:e00616. [PMID: 32608157 PMCID: PMC7327382 DOI: 10.1002/prp2.616] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
Microtubules (MTs) are highly dynamic polymers that constitute the cellular cytoskeleton and play a role in multiple cellular functions. Variability characterizes biological systems and is considered a part of the normal function of cells and organs. Variability contributes to cell plasticity and is a mechanism for overcoming errors in cellular level assembly and function, and potentially the whole organ level. Dynamic instability is a feature of biological variability that characterizes the function of MTs. The dynamic behavior of MTs constitutes the basis for multiple biological processes that contribute to cellular plasticity and the timing of cell signaling. Colchicine is a MT-modifying drug that exerts anti-inflammatory and anti-cancer effects. This review discusses some of the functions of colchicine and presents a platform for introducing variability while targeting MTs in intestinal cells, the microbiome, the gut, and the systemic immune system. This platform can be used for implementing novel therapies, improving response to chronic MT-based therapies, overcoming drug resistance, exerting gut-based systemic immune responses, and generating patient-tailored dynamic therapeutic regimens.
Collapse
Affiliation(s)
- Esther Forkosh
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Ariel Kenig
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| | - Yaron Ilan
- Department of MedicineHebrew University‐Hadassah Medical CentreJerusalemIsrael
| |
Collapse
|
123
|
Javier Díaz-García F, Flores-Medina S, Mercedes Soriano-Becerril D. Interplay between Human Intestinal Microbiota and Gut-to-Brain Axis: Relationship with Autism Spectrum Disorders. Microorganisms 2020. [DOI: 10.5772/intechopen.89998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
124
|
Wilhelm FR, Krautwald-Junghanns ME, Ortín-Piqueras V, Junnila J, Cramer K, Forsgård RA, Frias R, Spillmann T, Schmidt V. Iohexol-based measurement of intestinal permeability in birds. J Exot Pet Med 2020. [DOI: 10.1053/j.jepm.2020.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
125
|
Effects of antimicrobial peptide cathelicidin-BF on diarrhea controlling, immune responses, intestinal inflammation and intestinal barrier function in piglets with postweaning diarrhea. Int Immunopharmacol 2020; 85:106658. [PMID: 32531710 DOI: 10.1016/j.intimp.2020.106658] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
The objective was to evaluate the effects of antimicrobial peptide cathelicidin-BF (C-BF) treatment on diarrhea controlling, immune responses, intestinal inflammation, and intestinal barrier function in piglets with postweaning diarrhea. In this study, fifty-four weaned piglets with diarrhea were selected and treated with saline (control), C-BF or norfloxacin nicotinic (NFN) for 7 days. Here, we investigated the effects of C-BF on diarrhea controlling, growth performance, serum immune indicators, intestinal morphology, intestinal inflammation, and intestinal epithelial barrier function in the weaned piglets with diarrhea. The results showed that C-BF treatment decreased (P < 0.05) diarrheal index and increased (P < 0.05) average daily gain (ADG) and average daily feed intake (ADFI) compared with control group. C-BF treatment decreased (P < 0.05) levels of serum blood urea nitrogen (BUN) and aspartate aminotransferase (AST), but increased (P < 0.05) levels of serum A/G and alkaline phosphatase (ALP) compared with control group. The concentrations of IL-6, IL-8, tumor necrosis factor-α (TNF-α), and IgG were lower (P < 0.05), but IgA was greater (P < 0.05) for piglets treated by C-BF compared with those in control group. C-BF and NFN treatment decreased (P < 0.05) IL-6, IL-8, IL-22, IL-10 and transforming growth factor-β (TGF-β) production in the jejunum and ileum compared with the control group. C-BF treatment increased the expression levels of zonula occluden-1 (ZO-1), Occludin, and Claudin-1 in the jejunum and colon compared with the control group and the NFN group. In conclusion, these data indicate that C-BF treatment may be an effective therapeutic strategy for controlling post-weaning diarrhea, improving immune responses, attenuating intestinal inflammation and enhancing intestinal barrier function in piglets with postweaning diarrhea.
Collapse
|
126
|
Vuotto C, Battistini L, Caltagirone C, Borsellino G. Gut Microbiota and Disorders of the Central Nervous System. Neuroscientist 2020; 26:487-502. [PMID: 32441219 DOI: 10.1177/1073858420918826] [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] [Indexed: 12/13/2022]
Abstract
The gut microbiota, consisting of bacteria, fungi, archaea, viruses, and protozoa, together with their collective genomes (microbiome), plays a key role in immune system development and maturation, gut morphology, and in performing essential metabolic functions. Several factors, including lifestyle, body mass index, diet, antibiotic use, and the environment, influence the balance of the intestinal microbiota, whose alterations (the so-called dysbiosis) in recent years have been associated with the onset and/or progression of neurological and neuropsychiatric disorders. The purpose of this narrative review is to provide an overview of the possible involvement of the microbiota-gut-brain axis in the pathogenesis of diseases of the central nervous system, with a special focus on key issues and common misjudgments on the potential contribution of specific microorganisms.
Collapse
Affiliation(s)
- Claudia Vuotto
- Experimental Neuroscience, Santa Lucia Foundation IRCCS -Rome, Italy
| | - Luca Battistini
- Experimental Neuroscience, Santa Lucia Foundation IRCCS -Rome, Italy
| | - Carlo Caltagirone
- Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS -Rome, Italy
| | | |
Collapse
|
127
|
Nakada N, Mikami T, Horie K, Nagashio R, Sakurai Y, Sanoyama I, Yoshida T, Sada M, Kobayashi K, Sato Y, Okayasu I, Murakumo Y. Expression of CA2 and CA9 carbonic anhydrases in ulcerative colitis and ulcerative colitis-associated colorectal cancer. Pathol Int 2020; 70:523-532. [PMID: 32410301 DOI: 10.1111/pin.12949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Abstract
Ulcerative colitis (UC) is characterized by chronic inflammation in the colonic mucosa and submucosa with repeating relapse and remission, but the pathogenesis is unknown. Patients with long-standing UC are at high risk of neoplasm development. The aim of the present study was to identify molecules whose expression is associated with UC and UC-associated colorectal cancer (UCCA). Biopsy specimens from UC and normal colonic mucosae were analyzed using a proteomics approach, in which carbonic anhydrase 2 (CA2) was identified as a molecule downregulated in UC mucosae. Immunohistochemically, CA2 expression was detected in normal and diverticulitis mucosal epithelia, and its expression decreased as UC activity increased. CA2 expression was almost undetectable in UCCA. We also analyzed the expression of another carbonic anhydrase, CA9, and its upstream molecule, hypoxia-inducible factor-1α (HIF-1α), both of which are induced under hypoxic conditions. It was revealed that CA9 expression was relatively low in normal, diverticulitis and UC mucosae, and was upregulated in UCCA. HIF-1α expression was consistently low in all tissue types examined. In conclusion, these results suggest that CA2 and CA9 may be possible indicators of UC activity and UCCA development, respectively.
Collapse
Affiliation(s)
- Norihiro Nakada
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan.,Department of Pathology, Nakagami Hospital, Okinawa, Japan
| | - Tetuo Mikami
- Department of Pathology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Kayo Horie
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Aomori, Japan
| | - Ryo Nagashio
- Department of Molecular Diagnostics, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Yasutaka Sakurai
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Itaru Sanoyama
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tsutomu Yoshida
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Miwa Sada
- Department of Gastroenterology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kiyonori Kobayashi
- Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yuichi Sato
- Department of Molecular Diagnostics, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Isao Okayasu
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshiki Murakumo
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
128
|
Wellington MO, Hamonic K, Krone JEC, Htoo JK, Van Kessel AG, Columbus DA. Effect of dietary fiber and threonine content on intestinal barrier function in pigs challenged with either systemic E. coli lipopolysaccharide or enteric Salmonella Typhimurium. J Anim Sci Biotechnol 2020; 11:38. [PMID: 32318266 PMCID: PMC7158091 DOI: 10.1186/s40104-020-00444-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/09/2020] [Indexed: 01/10/2023] Open
Abstract
Background The independent and interactive effects of dietary fiber (DF) and threonine (Thr) were investigated in growing pigs challenged with either systemic E. coli lipopolysaccharide (LPS) or enteric Salmonella Typhimurium (ST) to characterise their effect on intestinal barrier function. Results In experiment 1, intestinal barrier function was assessed via oral lactulose and mannitol (L:M) gavage and fecal mucin analysis in pigs challenged with E. coli LPS and fed low fiber (LF) or high fiber (HF) diets with graded dietary Thr. Urinary lactulose recovery and L:M ratio increased (P < 0.05) during the LPS inoculation period in LF fed pigs but not in HF fed pigs. Fecal mucin output was increased (P < 0.05) in pigs fed HF compared to LF fed pigs. In experiment 2, RT-qPCR, ileal morphology, digesta volatile fatty acid (VFA) content, and fecal mucin output were measured in Salmonella Typhimurium challenged pigs, fed LF or HF diets with standard or supplemented dietary Thr. Salmonella inoculation increased (P < 0.05) fecal mucin output compared to the unchallenged period. Supplemental Thr increased fecal mucin output in the HF-fed pigs (Fib × Thr; P < 0.05). Feeding HF increased (P < 0.05) VFA concentration in cecum and colon. No effect of either Thr or fiber on expression of gene markers was observed except a tendency (P = 0.06) for increased MUC2 expression with the HF diet. Feeding HF increased goblet cell numbers (P < 0.05). Conclusion Dietary fiber appears to improve barrier function through increased mucin production capacity (i.e., goblet cell numbers, MUC2 gene expression) and secretion (i.e., fecal mucin output). The lack of effect of dietary Thr in Salmonella-challenged pigs provides further evidence that mucin secretion in the gut is conserved and, therefore, Thr may be limiting for growth under conditions of increased mucin production.
Collapse
Affiliation(s)
- Michael O Wellington
- 1Prairie Swine Centre, Inc., Saskatoon, SK S7H 5N9 Canada.,2Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8 Canada
| | - Kimberley Hamonic
- 2Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8 Canada
| | - Jack E C Krone
- 1Prairie Swine Centre, Inc., Saskatoon, SK S7H 5N9 Canada.,2Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8 Canada
| | - John K Htoo
- Evonik Nutrition & Care GmbH, Hanau-Wolfgang, Germany
| | - Andrew G Van Kessel
- 2Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8 Canada
| | - Daniel A Columbus
- 1Prairie Swine Centre, Inc., Saskatoon, SK S7H 5N9 Canada.,2Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK S7N 5A8 Canada
| |
Collapse
|
129
|
Peñalver Bernabé B, Maki PM, Dowty SM, Salas M, Cralle L, Shah Z, Gilbert JA. Precision medicine in perinatal depression in light of the human microbiome. Psychopharmacology (Berl) 2020; 237:915-941. [PMID: 32065252 DOI: 10.1007/s00213-019-05436-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/11/2019] [Indexed: 12/17/2022]
Abstract
Perinatal depression is the most common complication of pregnancy and affects the mother, fetus, and infant. Recent preclinical studies and a limited number of clinical studies have suggested an influence of the gut microbiome on the onset and course of mental health disorders. In this review, we examine the current state of knowledge regarding genetics, epigenetics, heritability, and neuro-immuno-endocrine systems biology in perinatal mood disorders, with a particular focus on the interaction between these factors and the gut microbiome, which is mediated via the gut-brain axis. We also provide an overview of experimental and analytical methods that are currently available to researchers interested in elucidating the influence of the gut microbiome on mental health disorders during pregnancy and postpartum.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States.
| | - Pauline M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shannon M Dowty
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Mariana Salas
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Lauren Cralle
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Zainab Shah
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Jack A Gilbert
- Scripts Oceanographic Institute, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
130
|
Qu F, Xu W, Deng Z, Xie Y, Tang J, Chen Z, Luo W, Xiong D, Zhao D, Fang J, Zhou Z, Liu Z. Fish c-Jun N-Terminal Kinase (JNK) Pathway Is Involved in Bacterial MDP-Induced Intestinal Inflammation. Front Immunol 2020; 11:459. [PMID: 32292404 PMCID: PMC7134542 DOI: 10.3389/fimmu.2020.00459] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/28/2020] [Indexed: 01/01/2023] Open
Abstract
The c-Jun NH2-terminal kinases (JNKs) are an evolutionarily conserved family of serine/threonine protein kinases that play critical roles in the pathological process in species ranging from insects to mammals. However, the function of JNKs in bacteria-induced intestinal inflammation is still poorly understood. In this study, a fish JNK (CiJNK) pathway was identified, and its potential roles in bacterial muramyl dipeptide (MDP)-induced intestinal inflammation were investigated in Ctenopharyngodon idella. The present CiJNK was found to possess a conserved dual phosphorylation motif (TPY) in a serine/threonine protein kinase (S_TKc) domain and to contain several potential immune-related transcription factor binding sites, including nuclear factor kappa B (NF-κB), activating protein 1 (AP-1), and signal transducer and activator of downstream transcription 3 (STAT3), in its 5′ flanking regions. Quantitative real-time PCR results revealed that the mRNA levels of the JNK pathway genes in the intestine were significantly upregulated after challenge with a bacterial pathogen (Aeromonas hydrophila) and MDP in a time-dependent manner. Additionally, the JNK pathway was found to be involved in regulating the MDP-induced expression levels of inflammatory cytokines (IL-6, IL-8, and TNF-α) in the intestine of grass carp. Moreover, the nutritional dipeptide carnosine and Ala–Gln could effectively alleviate MDP-induced intestinal inflammation by regulating the intestinal expression of JNK pathway genes and inflammatory cytokines in grass carp. Finally, fluorescence microscopy and dual-reporter assays indicated that CiJNK could associate with CiMKK4 and CiMKK7 involved in the regulation of the AP-1 signaling pathway. Overall, these results provide the first experimental demonstration that the JNK signaling pathway is involved in the intestinal immune response to MDP challenge in C. idella, which may provide new insight into the pathogenesis of inflammatory bowel disease.
Collapse
Affiliation(s)
- Fufa Qu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Wenqian Xu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Zhangren Deng
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Yifang Xie
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jianzhou Tang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Zhiguo Chen
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Wenjie Luo
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Ding Xiong
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Dafang Zhao
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Jiamei Fang
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China
| | - Zhigang Zhou
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhen Liu
- Hunan Provincial Key Laboratory of Nutrition and Quality Control of Aquatic Animals, Department of Biological and Environmental Engineering, Changsha University, Changsha, China.,Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
131
|
Li Q, Liang X, Xue X, Wang K, Wu L. Lipidomics Provides Novel Insights into Understanding the Bee Pollen Lipids Transepithelial Transport and Metabolism in Human Intestinal Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:907-917. [PMID: 31842537 DOI: 10.1021/acs.jafc.9b06531] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Bee pollen (BP) shows profound gut-protecting potentials. BP lipids (BPLs) mainly composed by phospholipids and polyunsaturated fatty acids might be one of the important contributors, while how BPL exerts gut-protecting effects and is transported through intestinal cell monolayers need to be investigated. Here, we exploited a strategy that combines an UPLC-Q-exactive orbitrap/MS-based lipidomics approach with a human intestinal cell (Caco-2) monolayer transport model, to determine the transepithelial transportation of BPL from Camellia sinensis L. (BPL-Cs), in pathological conditions. The results showed that BPL-Cs protected Caco-2 cells against dextran sulfate sodium (DSS)-induced intestinal barrier dysfunction by improving cell viability, maintaining membrane integrity, increasing tight junctions (ZO-1 and Claudin-1), and eliciting the expressions of antioxidative-related genes (NQO1, Nrf2, Txnrd1, and GSTA1). Lipidomics analysis revealed that DSS suppressed the transport and uptake of most of BPL-Cs including glycerophospholipids, sphingomyelins, and glycosylsphingolipids. Pretreatment with BPL-Cs significantly regulated glycerophospholipid and sphingolipid metabolisms, potentially involved in building permeability barriers and alleviating intestinal oxidative stress. Finally, eight classes of lipids were identified as the potential biomarkers for evaluating DSS-induced Caco-2 cell dysfunctions and BPL-intervened modulation. These findings shed light on the development of BPL as gastrointestinal protective food supplements in the future.
Collapse
Affiliation(s)
- Qiangqiang Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences , Beijing 100093 , China
| | - Xinwen Liang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences , Beijing 100093 , China
| | - Xiaofeng Xue
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences , Beijing 100093 , China
| | - Kai Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences , Beijing 100093 , China
| | - Liming Wu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences , Beijing 100093 , China
| |
Collapse
|
132
|
Carullo G, Governa P, Spizzirri UG, Biagi M, Sciubba F, Giorgi G, Loizzo MR, Di Cocco ME, Aiello F, Restuccia D. Sangiovese cv Pomace Seeds Extract-Fortified Kefir Exerts Anti-Inflammatory Activity in an In Vitro Model of Intestinal Epithelium Using Caco-2 Cells. Antioxidants (Basel) 2020; 9:E54. [PMID: 31936207 PMCID: PMC7022605 DOI: 10.3390/antiox9010054] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/04/2020] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease and food allergies are a growing topic in the field of nutrition science. Polyphenols, which are the most important secondary metabolites of plants, demonstrated to modulate the expression and/or production of numerous proteins, but also to regulate the intestinal ecosystem. In this context, our aim was the investigation of protective effects against the gastrointestinal mucosa of fortified milk kefir obtained by adding seeds extract from Sangiovese cv. Pomace. Methods: An ultrasound-assisted method was used to obtain the extracts. All the extracts were assayed for the antioxidant activity. The best extract was used as an additive of fermented milk kefir to obtain a fortified final product. Kefir samples were analyzed by NMR spectroscopy. The efficiency of the barrier functions was evaluated by measuring trans-epithelial electric resistance (TEER) using a voltmeter. Results: the enriched kefir (Ksgn) possesses higher antioxidant performances compared to the unfortified sample (Kwht). Kwht and Ksgn did not alter Caco-2 TEER in basal condition.
Collapse
Affiliation(s)
- Gabriele Carullo
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione—Dipartimento di Eccellenza 2018-2022, Università della Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; (G.C.); (U.G.S.); (M.R.L.); (D.R.)
| | - Paolo Governa
- Dipartimento di Biotecnologie, Chimica e Farmacia—Dipartimento di Eccellenza 2018-2022, Università di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (P.G.); (G.G.)
| | - Umile Gianfranco Spizzirri
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione—Dipartimento di Eccellenza 2018-2022, Università della Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; (G.C.); (U.G.S.); (M.R.L.); (D.R.)
| | - Marco Biagi
- Dipartimento di Scienze Fisiche, della Terra e dell’Ambiente, Università di Siena, Via Laterina 8, 53100 Siena, Italy;
| | - Fabio Sciubba
- Dipartimento di Chimica, Università di Roma “La Sapienza”, Piazzale Aldo Moro 5, 00185 Roma, Italy; (F.S.); (M.E.D.C.)
| | - Gianluca Giorgi
- Dipartimento di Biotecnologie, Chimica e Farmacia—Dipartimento di Eccellenza 2018-2022, Università di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (P.G.); (G.G.)
| | - Monica Rosa Loizzo
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione—Dipartimento di Eccellenza 2018-2022, Università della Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; (G.C.); (U.G.S.); (M.R.L.); (D.R.)
| | - Maria Enrica Di Cocco
- Dipartimento di Chimica, Università di Roma “La Sapienza”, Piazzale Aldo Moro 5, 00185 Roma, Italy; (F.S.); (M.E.D.C.)
| | - Francesca Aiello
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione—Dipartimento di Eccellenza 2018-2022, Università della Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; (G.C.); (U.G.S.); (M.R.L.); (D.R.)
| | - Donatella Restuccia
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione—Dipartimento di Eccellenza 2018-2022, Università della Calabria, Edificio Polifunzionale, 87036 Rende (CS), Italy; (G.C.); (U.G.S.); (M.R.L.); (D.R.)
| |
Collapse
|
133
|
Attenuation of DSS induced colitis by Dictyophora indusiata polysaccharide (DIP) via modulation of gut microbiota and inflammatory related signaling pathways. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103641] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
134
|
Alinaghi M, Nguyen DN, Sangild PT, Bertram HC. Direct Implementation of Intestinal Permeability Test in NMR Metabolomics for Simultaneous Biomarker Discovery-A Feasibility Study in a Preterm Piglet Model. Metabolites 2020; 10:metabo10010022. [PMID: 31906404 PMCID: PMC7022985 DOI: 10.3390/metabo10010022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/22/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022] Open
Abstract
Measurement of intestinal permeability (IP) is often used in the examination of inflammatory gastrointestinal disorders. IP can be assessed by measurement of urinary recovery of ingested non-metabolizable lactulose (L) and mannitol (M). The present study aimed to examine how measurements of IP can be integrated in a NMR-based metabolomics approach for a simultaneous quantification of L/M ratio and biomarker exploration. For this purpose, plasma and urine samples were collected from five-day-old preterm piglets (n = 20) with gastrointestinal disorders (subjected to intra-amniotic lipopolysaccharide (LPS, 1 mg/fetus)) after they had been administrated a 5% lactulose and 5% mannitol solution (15 mL/kg). The collected plasma and urine samples were analyzed by 1H NMR-based metabolomics. Urine L/M ratio measured by 1H NMR spectroscopy showed high correlation with the standard measurement of the urinary recoveries by enzymatic assays (r = 0.93, p < 0.05). Partial least squares (PLS) regressions and correlation analyses between L/M ratio and NMR metabolomics data revealed that L/M ratio was positively correlated with plasma lactate, acetate and succinate levels and negatively correlated with urinary hippuric acid and glycine. In conclusion, the present study demonstrated that NMR metabolomics enables simultaneous IP testing and discovery of biomarkers associated with an impaired intestinal permeability.
Collapse
Affiliation(s)
- Masoumeh Alinaghi
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev, Denmark;
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 68, 1860 Frederiksberg C, Denmark; (D.N.N.); (P.T.S.)
| | - Per Torp Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlaegevej 68, 1860 Frederiksberg C, Denmark; (D.N.N.); (P.T.S.)
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev, Denmark;
- Correspondence:
| |
Collapse
|
135
|
Ruff J, Barros TL, Tellez G, Blankenship J, Lester H, Graham BD, Selby CAM, Vuong CN, Dridi S, Greene ES, Hernandez-Velasco X, Hargis BM, Tellez-Isaias G. Research Note: Evaluation of a heat stress model to induce gastrointestinal leakage in broiler chickens. Poult Sci 2019; 99:1687-1692. [PMID: 32115037 PMCID: PMC7587818 DOI: 10.1016/j.psj.2019.10.075] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/25/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to evaluate heat stress as a model to induce gastrointestinal leakage in broiler chickens. On the day of hatch, 320 chicks were allocated into 8 environmental chambers, 4 thermoneutral (TN) and 4 continuous heat stress (HS). Each chamber was divided into 2 pens containing separate feeders and water jugs (8 replicates per treatment, 20 birds/pen). The environment was established to simulate production setting as best possible for the first 21 D. A gradual reduction of temperature from 32°C to 24°C with relative humidity at 55 ± 5% was adopted for the first 21 D. At the time of HS, the HS groups were exposed to 35°C from Day 21 to 42, while thermoneutral ones were maintained at 24°C from Day 21 to 42. Chickens were equipped with a Thermochron temperature logger for continuous monitoring of core body temperature. The environmental temperature and relative humidity were continuously recorded. Fluorescein isothiocyanate–dextran (FITC-d) was orally gavaged to 2 chickens/replicate (n = 16) randomly selected on days 21, 28, 35, and 42. After 1 h of oral gavage, blood samples were collected to determine the passage of FITC-d. Tibias were removed from all chickens to evaluate break strength only on 21 D and 42 D (before HS and at the end of the trial). Performance parameters were evaluated weekly from 21 D to the end of the trial. Body temperature was significantly (P < 0.05) increased after 2 h of starting HS and remained that way until the end of the study. Chronic HS caused an increase in core body temperature which decreased feed intake, body weight, and feed efficiency (28, 35, and 42 D) when compared with control TN chickens. Similarly, serum FITC-d was significantly increased in HS chickens at all points of evaluation. Chronic HS also caused a significant reduction of bone strength at 42 D when compared with the control chickens. The results from the present study suggest that HS can be a robust model to induce gut leakage in broiler chickens.
Collapse
Affiliation(s)
- Jared Ruff
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Thaina L Barros
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Guillermo Tellez
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Justin Blankenship
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Howard Lester
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Brittany D Graham
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Callie A M Selby
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Christine N Vuong
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Sami Dridi
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth S Greene
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - X Hernandez-Velasco
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico 04510, Mexico
| | - Billy M Hargis
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | | |
Collapse
|
136
|
Qin SM, Zhang KY, Ding XM, Bai SP, Wang JP, Zeng QF. Effect of dietary graded resistant potato starch levels on growth performance, plasma cytokines concentration, and intestinal health in meat ducks. Poult Sci 2019; 98:3523-3532. [PMID: 31329991 DOI: 10.3382/ps/pez186] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
The objective of the present study was to investigate the effect of dietary graded raw potato starch (RPS) levels on growth performance, plasma cytokines concentration, ileal barrier function, and cecal short-chain fatty acids (SCFA) concentration in meat ducks from 1 to 35 D of age. This study included 2 experiments. In experiment (Exp.) 1, sixteen 35-day-old meat ducks were used to evaluate the AME of RPS by orogastric administration. Results showed the AME value of RPS on ducks is 2.76 kcal/g. In Exp. 2, a total of 600 one-day-old ducklings were randomly assigned to 5 isonitrogenous and isoenergetic dietary treatments that included 0 (control), 6, 12, 18, and 24% RPS, respectively. Samples were collected at both of 14 and 35 D. Neither growth performance nor ileal parameters (length, weight, and pH) at both of 14 and 35 D was affected by dietary RPS. However, the mucosal thickness (14 D), villus height (except for 18% RPS at 14 D), and the villus height: crypt depth ratio (14 and 35 D) of the ileum were increased in the 12 and 18% RPS diets when compared to 0% RPS diet. Meanwhile, proinflammatory factors such as plasma interleukin (IL)-1β and IL-6 (14 D) reduced in 12% RPS diet and tumor necrosis factor α decreased in 12% (except for 14 D) and 18% RPS groups. When compared with the control group, diets with 18% RPS significantly increased mucin 2 gene expression at 14 D, and 12% RPS elevated the mRNA expression of tight junction proteins including Zonula occludens-1 and Claudin 1 (except for 14 D) in the ileal mucosa of birds. Furthermore, ducks fed 12% RPS diet had higher concentrations of acetate, propionate, and butyrate in cecal digesta than other groups. These findings indicated that diets with 12 and/or 18% RPS increased the cecal SCFA concentration, which subsequently enhanced the barrier function and improved intestinal health in the ileum for 14 and 35-day-old meat ducks.
Collapse
Affiliation(s)
- S M Qin
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - K Y Zhang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - X M Ding
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - S P Bai
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - J P Wang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Q F Zeng
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
137
|
Grochowska M, Laskus T, Radkowski M. Gut Microbiota in Neurological Disorders. Arch Immunol Ther Exp (Warsz) 2019; 67:375-383. [PMID: 31578596 PMCID: PMC6805802 DOI: 10.1007/s00005-019-00561-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
The incidence of neurological disorders such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD) is increasing throughout the world, but their pathogenesis remains unclear and successful treatment remains elusive. Bidirectional communications between the central nervous system and gut microbiota may play some role in the pathogenesis of the above disorders. Up to a thousand bacterial species reside in human intestine; they colonize the gut shortly after birth and remain for life. Numerous studies point to the role of microbiota composition in the development, course and treatment of MS, AD and PD.
Collapse
Affiliation(s)
- Marta Grochowska
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, Warsaw, Poland.
| | - Tomasz Laskus
- Department of Adult Infectious Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Marek Radkowski
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
138
|
Dong Y, Qu X, Wu G, Luo X, Tang B, Wu F, Fan L, Dev S, Liang T. Advances in the Detection, Mechanism and Therapy of Chronic Kidney Disease. Curr Pharm Des 2019; 25:4235-4250. [PMID: 31742493 DOI: 10.2174/1381612825666191119094354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 10/30/2019] [Indexed: 01/08/2023]
Abstract
Chronic Kidney Disease (CKD) is characterized by the gradual loss of renal mass and functions. It has become a global health problem, with hundreds of millions of people being affected. Both its incidence and prevalence are increasing over time. More than $20,000 are spent on each patient per year. The economic burden on the patients, as well as the society, is heavy and their life quality worsen over time. However, there are still limited effective therapeutic strategies for CKD. Patients mainly rely on dialysis and renal transplantation, which cannot prevent all the complications of CKD. Great efforts are needed in understanding the nature of CKD progression as well as developing effective therapeutic methods, including pharmacological agents. This paper reviews three aspects in the research of CKD that may show great interests to those who devote to bioanalysis, biomedicine and drug development, including important endogenous biomarkers quantification, mechanisms underlying CKD progression and current status of CKD therapy.
Collapse
Affiliation(s)
- Yu Dong
- Department of Urology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 530011, Nanning, China
| | - Xiaosheng Qu
- National Engineering Laboratory of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, No. 189, Changgang Road, 530023, Nanning, China
| | - Gang Wu
- Department of Urology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 530011, Nanning, China
| | - Xiangdong Luo
- Department of Urology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 530011, Nanning, China
| | - Botao Tang
- Department of Urology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 530011, Nanning, China
| | - Fangfang Wu
- National Engineering Laboratory of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, No. 189, Changgang Road, 530023, Nanning, China
| | - Lanlan Fan
- School of Pharmacy, Guangxi University of Chinese Medicine, 530001, Nanning, China
| | - Sooranna Dev
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, 369, Fulham Road, London SW10 9NH, United Kingdom
| | - Taisheng Liang
- Department of Urology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 530011, Nanning, China
| |
Collapse
|
139
|
Takeshita M, Tanaka A, Nakamura T, Sato E, Node K. Effect of Lubiprostone on Urinary Protein Excretion: A Report of Two IgA Nephropathy Patients with Chronic Constipation. Intern Med 2019; 58:3255-3259. [PMID: 31292404 PMCID: PMC6911766 DOI: 10.2169/internalmedicine.3158-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Disturbance of the normal gut microbiota has been implicated in the pathogenesis of various diseases, including chronic kidney disease (CKD). A common CKD symptom is chronic constipation. Lubiprostone is a safe and efficacious drug for treating chronic constipation. We herein report 2 patients with IgA nephropathy treated with lubiprostone (24 μg 1×/day). The lubiprostone treatment ameliorated their chronic constipation and, unexpectedly, reduced the urinary protein excretion, urinary liver-type fatty acid binding protein and urine occult blood. These results may indicate that lubiprostone is a useful therapeutic intervention against the progression of IgA nephropathy with chronic constipation.
Collapse
Affiliation(s)
| | - Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, Japan
| | - Tsukasa Nakamura
- Division of Nephrology, Department of Internal Medicine, Shinmatsudo Central General Hospital, Japan
| | - Eiichi Sato
- Division of Nephrology, Department of Internal Medicine, Shinmatsudo Central General Hospital, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Japan
| |
Collapse
|
140
|
Jin C, Zeng Z, Wang C, Luo T, Wang S, Zhou J, Ni Y, Fu Z, Jin Y. Insights into a Possible Mechanism Underlying the Connection of Carbendazim-Induced Lipid Metabolism Disorder and Gut Microbiota Dysbiosis in Mice. Toxicol Sci 2019; 166:382-393. [PMID: 30496565 DOI: 10.1093/toxsci/kfy205] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Carbendazim (CBZ), a systemic, broad-spectrum benzimidazole fungicide, is widely used to control fungal diseases and has been regarded as an endocrine disruptor that causes mammalian toxicity in different target organs. Here, we discovered that chronic administrations of CBZ at 0.2, 1, and 5 mg/kg body weight for 14 weeks not only changed the composition of gut microbiota but also induced significant increases in body, liver, and epididymal fat weight in mice. At the biochemical level, the serum triglyceride (TG) and glucose levels also increased after CBZ exposure. Moreover, the level of serum lipoprotein lipase (LPL), which plays an important role in fatty acid release from TG, was decreased significantly. For gut microbiota, 16S rRNA gene sequencing and real-time qPCR revealed that CBZ exposure significantly perturbed the mice gut microbiome, and gas chromatography found that the production of short-chain fatty acids were altered. Moreover, CBZ exposure increased the absorption of exogenous TG in the mice intestine and inhibited the TG consumption, eventually leading the serum triglyceride to maintain higher levels. The increase of lipid absorption in the intestine direct caused hyperlipidemia and the multi-tissue inflammatory response. In response to the rise of lipid in blood, the body maintains the balance of lipid metabolism in mice by reducing lipid synthesis in the liver and increasing lipid storage in the fat. Chronic CBZ exposure induced the gut microbiota dysbiosis and disturbed lipid metabolism, which promoted the intestinal absorption of excess triglyceride and caused multiple tissue inflammatory responses in mice.
Collapse
Affiliation(s)
- Cuiyuan Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhaoyang Zeng
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Caiyun Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ting Luo
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Siyu Wang
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jicong Zhou
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yingchun Ni
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhengwei Fu
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yuanxiang Jin
- Department of Biotechnology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
141
|
Wei HC, Chen P, Liang XF, Yu HH, Wu XF, Han J, Luo L, Gu X, Xue M. Plant protein diet suppressed immune function by inhibiting spiral valve intestinal mucosal barrier integrity, anti-oxidation, apoptosis, autophagy and proliferation responses in amur sturgeon (Acipenser schrenckii). FISH & SHELLFISH IMMUNOLOGY 2019; 94:711-722. [PMID: 31574297 DOI: 10.1016/j.fsi.2019.09.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 06/10/2023]
Abstract
An 8-week growth trial was conducted to investigate the effects of replacing dietary fishmeal with a plant protein blend on the growth performance, mucosal barrier integrity and the related regulation mechanism in Amur Sturgeon (Acipenser schrenckii) with initial weight of 87.48 g. Three isonitrogenous and isoenergetic diets were prepared. A basal diet containing 540 g/kg fishmeal (P0), whereas the other two diets were formulated by replacing 50% and 100% of FM with plant protein blend (soybean protein concentrate and cottonseed protein concentrate), and named as P50 and P100, respectively. Although essential amino acids, fatty acids, and available phosphorus had been balanced according to the nutrient requirement of sturgeon, compared with the fish of P0 and P50, the full plant protein diet (P100) significantly reduced growth performance and survival, and accompanied with serious spiral valve intestinal (SVI) damage. The increased tissue necrosis and failed responses in anti-oxidation, programming apoptosis, autophagy and cell proliferation system were regulated by inhibiting ERK1 phosphorylation, which indicated that SVI hypoimmunity and functional degradation were the main reasons for the high mortality and low utilization ability of plant protein in Amur sturgeon.
Collapse
Affiliation(s)
- H C Wei
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - P Chen
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - X F Liang
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - H H Yu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Beijing Fisheries Research Institute, Beijing, 100068, China
| | - X F Wu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - J Han
- Institute of Food and Nutrition Development, Ministry of Agriculture, Beijing, 100081, China
| | - L Luo
- Beijing Fisheries Research Institute, Beijing, 100068, China
| | - X Gu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Agriculture and Rural Ministry Quality and Safety Risk Evaluation Laboratory of Feed and Feed Additives for Animal Husbandry, Beijing, 100081, China.
| | - M Xue
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Agriculture and Rural Ministry Quality and Safety Risk Evaluation Laboratory of Feed and Feed Additives for Animal Husbandry, Beijing, 100081, China.
| |
Collapse
|
142
|
Pan Z, Yuan X, Tu W, Fu Z, Jin Y. Subchronic exposure of environmentally relevant concentrations of F-53B in mice resulted in gut barrier dysfunction and colonic inflammation in a sex-independent manner. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 253:268-277. [PMID: 31319243 DOI: 10.1016/j.envpol.2019.07.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/25/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
F-53B (6:2 chlorinated polyfluorinated ether sulfonate) is currently recognized as a safe alternative to long-chain PFASs in China. However, an increasing number of studies have recently authenticated its biotoxicological effects. In this study, for evaluating the gut toxicity of F-53B in mammals, both female and male mice were orally exposed to 0, 1, 3, or 10 μg/L F-53B for 10 weeks. Our results showed that F-53B significantly accumulated in the colon, ileum and serum when exposed to 10 μg/L F-53B for 10 weeks. F-53B exposure not only increased the transcriptional levels of ion transport-related genes but could also interact with the CFTR protein directly. Interestingly, subchronic F-53B exposure also increased the transcription of mucus secretion-related genes, but the protein level of Muc2 decreased after F-53B exposure, indicating that there was a compensatory phenomenon after mucus barrier injury. Furthermore, F-53B exposure also induced colonic inflammation associated with gut microbiota dysbiosis in the colon. Taken together, our results indicated that the potential gut toxicity of F-53B and almost all of the changed parameters were significantly affected in both female and male mice, suggesting that F-53B could disturb the gut barrier without sex dependence in mice.
Collapse
Affiliation(s)
- Zihong Pan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xianling Yuan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang, 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China.
| |
Collapse
|
143
|
Salguero MV, Al-Obaide MAI, Singh R, Siepmann T, Vasylyeva TL. Dysbiosis of Gram-negative gut microbiota and the associated serum lipopolysaccharide exacerbates inflammation in type 2 diabetic patients with chronic kidney disease. Exp Ther Med 2019; 18:3461-3469. [PMID: 31602221 PMCID: PMC6777309 DOI: 10.3892/etm.2019.7943] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/29/2019] [Indexed: 12/11/2022] Open
Abstract
Lipopolysaccharide (LPS), a potent endotoxin present in the outer membrane of Gram-negative bacteria, causes chronic immune responses associated with inflammation. In the present study, the association between LPS and the dysbiosis of Gram-negative bacteria in the gut microbiome was determined in patients with type 2 diabetes mellitus (T2DM) and chronic kidney disease (T2DM-CKD; stages 4 and 5, not on dialysis) compared with healthy individuals. Microbiome diversity was analyzed in patients with T2DM-CKD and healthy controls by sequencing the hypervariable sub-regions of the 16S ribosomal RNA gene from stool samples. Serum samples were assayed by ELISA for LPS, C-reactive protein (CRP), tumor necrosis factor-α (TNFα), interleukin-6 (IL6) and endothelin-1. A total of four gut Gram-negative phyla (Bacteroidetes, Proteobacteria, Fusobacteria and Verrucomicrobia) were identified in the gut microbiome of the T2DM-CKD and control groups. Proteobacteria, Verrucomicrobia and Fusobacteria exhibited significantly increased relative abundance in patients with T2DM-CKD when compared with controls (P<0.05). The levels of serum LPS were significantly increased in patients with T2DM-CKD compared with controls (P<0.05). Elevated serum LPS was significantly correlated with increased levels of TNFα, IL6 and CRP. The dysbiosis of Gram-negative bacteria in the gut microbiome of patients with T2DM-CKD may contribute to the elevated serum levels of LPS and the consequential effects on the inflammatory biomarkers in these patients. The association between the dysbiosis of Gram-negative bacteria in the gut microbiome of patients with T2DM-CKD, increased LPS levels and the effects on inflammatory biomarkers may provide insight into potential diagnostic and therapeutic approaches in the treatment of T2DM-CKD.
Collapse
Affiliation(s)
- Maria V Salguero
- Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.,Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Saxony D-01067, Germany
| | - Mohammed A I Al-Obaide
- Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ruchi Singh
- Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Timo Siepmann
- Center for Clinical Research and Management Education, Division of Health Care Sciences, Dresden International University, Dresden, Saxony D-01067, Germany.,Department of Neurology, University Hospital Carl Gustav Carus, Technische Universitat Dresden, Dresden, Saxony D-01307, Germany
| | - Tetyana L Vasylyeva
- Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
144
|
Antonini M, Lo Conte M, Sorini C, Falcone M. How the Interplay Between the Commensal Microbiota, Gut Barrier Integrity, and Mucosal Immunity Regulates Brain Autoimmunity. Front Immunol 2019; 10:1937. [PMID: 31475000 PMCID: PMC6706873 DOI: 10.3389/fimmu.2019.01937] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022] Open
Abstract
The intestinal barrier provides the host with a strong defense line against the external environment playing also a pivotal role in the crosstalk between the gut microbiota and the immune system. Notably, increasing lines of evidence concerning autoimmune disorders such as Multiple Sclerosis (MS) report an imbalance in both intestinal microbiota composition and mucosal immunity activation, along with an alteration of gut barrier permeability, suggesting this complex network plays a crucial role in modulating the course of autoimmune responses occurring in tissues outside the gut such as the central nervous system (CNS). Here, we review current knowledge on how gut inflammation and breakage of gut barrier integrity modulates the interplay between the commensal gut microbiota and the immune system and its role in shaping brain immunity.
Collapse
Affiliation(s)
- Martina Antonini
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Lo Conte
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Sorini
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Marika Falcone
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
145
|
Dreyer L, Smith C, Deane SM, Dicks LMT, van Staden AD. Migration of Bacteriocins Across Gastrointestinal Epithelial and Vascular Endothelial Cells, as Determined Using In Vitro Simulations. Sci Rep 2019; 9:11481. [PMID: 31391488 PMCID: PMC6685951 DOI: 10.1038/s41598-019-47843-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/24/2019] [Indexed: 01/18/2023] Open
Abstract
Little is known about the migration of bacteriocins across human cells. In this study, we report on migration of three bacteriocins nisin, plantaricin 423 and bacST4SA across colonic adenocarcinoma (Caco-2) cells and human umbilical vein endothelial cells (HUVECs). Bacteriocins were fluorescently labelled while still maintaining antimicrobial activity. Migration of fluorescently labelled bacteriocins across monolayers was assessed in vitro using transmigration well inserts. After 3 h, 75% of nisin, 85% of plantaricin 423 and 82% of bacST4SA migrated across the Caco-2 cell monolayer. Over the same time span, 88% nisin, 93% plantaricin 423 and 91% bacST4SA migrated across the HUVEC monolayer. The viability of both cell types remained unchanged when exposed to 50 µM of nisin, plantaricin 423 or bacST4SA. The effect of human plasma on bacteriocin activity was also assessed. Activity loss was dependent on bacteriocin type and concentration, with the class-IIa bacteriocins retaining more activity compared to nisin. This is the first report of bacteriocins migrating across simulated gastrointestinal- and vascular-barriers. This study provides some of the first evidence that bacteriocins are capable of crossing the gut-blood-barrier. However, in vivo studies need to be performed to confirm these findings and expand on the role of bacteriocin migration across cell barriers.
Collapse
Affiliation(s)
- Leané Dreyer
- Department of Microbiology, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Shelly M Deane
- Department of Microbiology, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, 7600, South Africa.
| | - Anton D van Staden
- Department of Microbiology, Stellenbosch University, Stellenbosch, 7600, South Africa.
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa.
| |
Collapse
|
146
|
Wang C, Zhang Y, Deng M, Wang X, Tu W, Fu Z, Jin Y. Bioaccumulation in the gut and liver causes gut barrier dysfunction and hepatic metabolism disorder in mice after exposure to low doses of OBS. ENVIRONMENT INTERNATIONAL 2019; 129:279-290. [PMID: 31146162 DOI: 10.1016/j.envint.2019.05.056] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 05/23/2023]
Abstract
The compound sodium ρ-perfluorous nonenoxybenzene sulfonate (OBS), a new kind of perfluoroalkyl and polyfluoroalkyl compound, is a surfactant for increasing oil production, and it has been widely detected in various organisms. Because of its wide use, OBS is detectable in the environment. However, knowledge about the biological toxicity of OBS to animals is very limited. Here, male mice were exposed to 0, 0.1, 1 or 10 μg/L of OBS for 6 weeks via drinking water. It was demonstrated that OBS was highly bioaccumulated both in the liver and gut in the mice after low doses of OBS exposure. Curiously, a low dose of OBS exposure also caused gut barrier dysfunction by decreasing mucus secretion and altering Ionic transport in the gut via the CFTR pathway. In addition, liver function was influenced by OBS at both the histopathological and physiological levels. Hepatic transcriptomics and metabolomics analysis showed a total of 1157 genes, and multiple metabolites changed significantly in the livers of mice exposed to low-dose OBS for 6 weeks. The functions of these changed genes and metabolites are tightly related to glycolysis, fatty acid synthesis, fatty acid transport, and β-oxidation. All these results indicate that the liver and gut are important target tissues for OBS exposure. Importantly, it is possible that high levels of bioaccumulation of OBS in the gut and liver might directly cause gut barrier dysfunction and hepatic metabolism disorder in mice.
Collapse
Affiliation(s)
- Caiyun Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yi Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Mi Deng
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China
| | - Xia Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wenqing Tu
- Research Institute of Poyang Lake, Jiangxi Academy of Sciences, Nanchang 330029, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
147
|
Lu R, Zhang YG, Xia Y, Sun J. Imbalance of autophagy and apoptosis in intestinal epithelium lacking the vitamin D receptor. FASEB J 2019; 33:11845-11856. [PMID: 31361973 DOI: 10.1096/fj.201900727r] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis and autophagy are dynamic processes that determine the fate of cells. Vitamin D receptor (VDR) deficiency in the intestine leads to abnormal Paneth cells and impaired autophagy function. Here, we will elucidate the mechanisms of the intestinal epithelial VDR regulation of autophagy and apoptosis. We used in vivo VDRlox and VDR∆IEC mice and ex vivo organoids generated from small intestine and colon tissues. We found that VDR deficiency induced more apoptotic cells and significantly increased cell death in the small intestine and colon of VDR∆IEC mice. The proapoptotic protein B-cell lymphoma 2 (BCL-2) associated X protein (Bax) was enhanced, whereas autophagy related 16 like 1 (ATG16L1) and Beclin-1 were decreased in the intestines of VDRΔIEC mice. Apoptosis induced by Bax reduced autophagy by decreasing Beclin-1. Physical interactions between Beclin-1 and Bcl-2 were increased in the VDR-deficient epithelia from mice. The growth of VDR∆IEC organoids was significantly slower with fewer Paneth cells than that of VDR+/+ organoids. The expression levels of Beclin-1 and lysozyme were decreased in VDR∆IEC organoids. Bacterial endotoxin levels were high in the serum from VDR∆IEC mice and made mice susceptible to colitis. In the organoids and colitis IL-10-/- mice, vitamin D3 treatment increased VDR and ATG16L1 protein expression levels, which activated autophagic responses. In summary, intestinal epithelial VDR regulates autophagy and apoptosis through ATG16L1 and Beclin-1. Our studies provide fundamental insights into the tissue-specific function of VDR in modulating the balance between autophagy and apoptosis.-Lu, R., Zhang, Y.-G., Xia, Y., Sun, J. Imbalance of autophagy and apoptosis in intestinal epithelium lacking the vitamin D receptor.
Collapse
Affiliation(s)
- Rong Lu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yong-Guo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yinglin Xia
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, USA.,University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
148
|
Jaworska K, Konop M, Bielinska K, Hutsch T, Dziekiewicz M, Banaszkiewicz A, Ufnal M. Inflammatory bowel disease is associated with increased gut-to-blood penetration of short-chain fatty acids: A new, non-invasive marker of a functional intestinal lesion. Exp Physiol 2019; 104:1226-1236. [PMID: 31243807 DOI: 10.1113/ep087773] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/29/2019] [Indexed: 01/02/2023]
Abstract
NEW FINDINGS What is the central question of this study? 'Leaky gut' has been found in intestinal and extra-intestinal diseases. However, functional evaluation of intestinal permeability is not widely used as a diagnostic marker, possibly owing to significant limitations of currently used permeability assays. There is an unmet need for development of a new, non-invasive test to assess intestinal function. What is the main finding and its importance? We show that an increased blood-to-stool ratio of the concentration of gut bacteria-produced short-chain fatty acids may be used as a marker of gut permeability. Our findings lay the groundwork for establishing a new, non-invasive, risk-free diagnostic tool in diseases associated with intestinal barrier malfunction, such as inflammatory bowel disease. ABSTRACT Intestinal diseases, such as inflammatory bowel disease (IBD), are characterized by an impaired gut-blood barrier commonly referred to as 'leaky gut'. Therefore, functional evaluation of the gut-blood barrier is a promising diagnostic marker. We hypothesized that short-chain fatty acids (SCFAs) produced by gut bacteria might serve as a marker in IBD. Animal experiments were performed on male Sprague-Dawley rats with acetic acid-induced colitis and in sham control animals. The gut-blood barrier permeability was determined by assessing the ratios of the following: (i) portal blood concentration of SCFAs (Cp ) to faecal concentration of SCFAs (Cf ); (ii) systemic blood concentration of SCFAs (Cs ) to faecal concentration of SCFAs (Cf ); and (iii) Cp and Cs of fluorescein isothiocyanate (FITC)-dextran administered into the colon. As a clinical study, we evaluated Cs , Cf and the Cs /Cf ratio of SCFAs in six paediatric patients with IBD, assessed as mild/moderate/severe by the Paediatric Ulcerative Colitis Activity Index (PUCAI) and the Paediatric Crohn's Disease Activity Index (PCDAI) at the time of sample collection, and nine age-matched healthy control subjects. Rats with histologically confirmed IBD had significantly increased ratios of Cp /Cf and Cs /Cf for SCFAs. This was positively correlated with the plasma FITC-dextran concentration. Likewise, IBD patients showed a significantly higher Cs /Cf ratio for SCFAs, including acetic, valeric, isocaproic, caproic and propionic acids, in comparison to control subjects. In conclusion, in the rats and in paediatric patients with IBD we found an increased blood-to-stool ratio of SCFAs, suggesting an increased gut-to-blood penetration of SCFAs. These findings pave the way for a new, non-invasive diagnostic tool in IBD and other diseases accompanied by intestinal barrier malfunction.
Collapse
Affiliation(s)
- Kinga Jaworska
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Marek Konop
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia Bielinska
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Hutsch
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Dziekiewicz
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Banaszkiewicz
- Department of Pediatric Gastroenterology and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
149
|
Qi X, Zhang Y, Guo H, Hai Y, Luo Y, Yue T. Mechanism and intervention measures of iron side effects on the intestine. Crit Rev Food Sci Nutr 2019; 60:2113-2125. [PMID: 31232087 DOI: 10.1080/10408398.2019.1630599] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Excess oral iron in the intestinal tract usually produces reactive oxygen species via Fenton and Haber-Weiss reaction, so oxidative stress is triggered. Lipid peroxidation procedurally appears, ferroptosis, apoptosis and necrosis are often induced, subsequently, mitochondrial damage, endoplasmic reticulum dysfunction and even cell death occur. As a result, the intestinal epithelial cells are destroyed, leading to the incompleteness of intestinal mechanical barrier. Simultaneously, iron supplement can change the compositions and metabolic processes of intestinal microbes, and the intestinal inflammatory may be worsened. In principle, the easier dissociation of Fe2+ from oral iron supplements is, the more serious intestinal inflammation will occur. Fortunately, some interventions have been developed to alleviate these side effects. For instance, some antioxidants e.g. VE and ferulic acid have been used to prevent the formation of free radicals or to neutralize the formed free radicals. Furthermore, some new iron supplements with the ability of slow-releasing Fe2+, e.g. ferrous citrate liposome and EDTA iron sodium, have been successfully prepared. In order to recover the intestinal micro-ecological balance, probiotics and prebiotics, bacterial consortium transplantation, and fecal microbiota transplantation have been developed. This study is meaningful for us to develop safer oral iron supplements and to maintain intestinal micro-ecological health.
Collapse
Affiliation(s)
- Xiao Qi
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi, China.,School of Chemical Engineering, Northwest University, Xi'an, Shaanxi, China
| | - Yuanxiao Zhang
- School of Chemical Engineering, Northwest University, Xi'an, Shaanxi, China
| | - Hang Guo
- School of Chemical Engineering, Northwest University, Xi'an, Shaanxi, China
| | - Yu Hai
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi, China
| | - Yane Luo
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi, China
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi, China.,Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Agro-products (Yangling), Beijing, China
| |
Collapse
|
150
|
Hu DJK, Jasper H. Epithelia: Understanding the Cell Biology of Intestinal Barrier Dysfunction. Curr Biol 2019; 27:R185-R187. [PMID: 28267974 DOI: 10.1016/j.cub.2017.01.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Barrier dysfunction in the intestine is a common characteristic of aging organisms. A recent study provides new insight into the cell biology of this phenomenon.
Collapse
Affiliation(s)
- Daniel J-K Hu
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|