1
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023:10.1007/s13311-023-01435-8. [PMID: 37759139 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
2
|
Martinez J, Rodriguez Hovnanian KM, Martinez EE. Biomarkers and Functional Assays of Epithelial Barrier Disruption and Gastrointestinal Dysmotility in Critical Illness-A Narrative Review. Nutrients 2023; 15:4052. [PMID: 37764835 PMCID: PMC10535972 DOI: 10.3390/nu15184052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Enteral nutrition in critically ill children has been associated with improved clinical outcomes. Gastrointestinal dysfunction often impedes the timely initiation and advancement of enteral nutrition and can contribute to immune dysregulation and systemic inflammation. Therefore, assessing gastrointestinal function, at a cellular and functional level, is important to provide optimal enteral nutrition therapy and reduce the gastrointestinal tract's contribution to the inflammatory cascade of critical illness. In this narrative review, we present an overview of biomarker and functional assays for gastrointestinal dysfunction, including epithelial barrier disruption and gastrointestinal dysmotility, that have been considered for critically ill patients.
Collapse
Affiliation(s)
- Julianna Martinez
- Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - K Marco Rodriguez Hovnanian
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Enid E Martinez
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
3
|
Wang Y, Chang HW, Lee EM, Zhou C, Pruss KM, Henrissat S, Chen RY, Kao C, Hibberd MC, Lynn HM, Webber DM, Crane M, Cheng J, Rodionov DA, Arzamasov AA, Castillo JJ, Couture G, Chen Y, Balcazo NP, Lebrilla CB, Terrapon N, Henrissat B, Ilkayeva O, Muehlbauer MJ, Newgard CB, Mostafa I, Das S, Mahfuz M, Osterman AL, Barratt MJ, Ahmed T, Gordon JI. Prevotella copri -linked effects of a therapeutic food for malnutrition. bioRxiv 2023:2023.08.11.553030. [PMID: 37645712 PMCID: PMC10461977 DOI: 10.1101/2023.08.11.553030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Preclinical and clinical studies are providing evidence that the healthy growth of infants and children reflects, in part, healthy development of their gut microbiomes 1-5 . This process of microbial community assembly and functional maturation is perturbed in children with acute malnutrition. Gnotobiotic animals, colonized with microbial communities from children with severe and moderate acute malnutrition, have been used to develop microbiome-directed complementary food (MDCF) formulations for repairing the microbiomes of these children during the weaning period 5 . In the accompanying paper 1 , we analyze bacterial genomes assembled from sequencing the fecal microbiomes of Bangladeshi children with moderate acute malnutrition (MAM) who had participated in a previously reported 3-month randomized controlled clinical study of an MDCF 6 . This formulation, MDCF-2, produced significantly improved weight gain compared to a commonly used nutritional intervention despite the lower caloric density of the MDCF. Characterizing the metagenome assembled genomes (MAGs) of bacterial strains present in the microbiomes of study participants revealed a significant correlation between accelerated ponderal growth and the expression by two Prevotella copri MAGs of metabolic pathways involved in processing of MDCF-2 glycans 1 . To provide a direct test of these relationships, in this report we perform 'reverse translation' experiments. These experiments center on a gnotobiotic mouse model of mother-to-offspring microbiome transmission involving sequential introduction of defined consortia of age- and ponderal growth-associated gut bacterial strains cultured from Bangladeshi infants/children in the study population, with or without P. copri isolates resembling the MAGs. By combining analyses of microbial community assembly, gene expression and processing of glycan constituents of MDCF-2 with single nucleus RNA-Seq and mass spectrometric analyses of the intestine, we establish a key role for the P. copri strains in mediating metabolism of MDCF-2 glycans, as well as the activities of metabolic pathways involved in lipid, amino acid, carbohydrate plus other facets of energy metabolism within epithelial cells positioned at different locations in intestinal crypts and villi. Together, the results described in these two papers provide insights into structure/function relationships between MDCF-2 and members of the gut communities of malnourished children; they also have implications for developing future prebiotic, probiotic and/or synbiotic therapeutics for microbiome restoration in children with already manifest malnutrition, or who are at risk for this pervasive health challenge.
Collapse
Affiliation(s)
- Yi Wang
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Hao-Wei Chang
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Evan M. Lee
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Cyrus Zhou
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Kali M. Pruss
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Suzanne Henrissat
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, F-13288, Marseille, France
| | - Robert Y. Chen
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Clara Kao
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Matthew C. Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Hannah M. Lynn
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Daniel M. Webber
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marie Crane
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Jiye Cheng
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Juan J. Castillo
- Department of Chemistry, University of California, Davis, CA 95616 USA
| | - Garret Couture
- Department of Chemistry, University of California, Davis, CA 95616 USA
| | - Ye Chen
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Chemistry, University of California, Davis, CA 95616 USA
| | - Nikita P. Balcazo
- Department of Chemistry, University of California, Davis, CA 95616 USA
| | | | - Nicolas Terrapon
- Architecture et Fonction des Macromolécules Biologiques, CNRS, Aix-Marseille University, F-13288, Marseille, France
| | - Bernard Henrissat
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710 USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710 USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710 USA
| | - Michael J. Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710 USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710 USA
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710 USA
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710 USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710 USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Subhasish Das
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Mustafa Mahfuz
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Andrei L. Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Michael J. Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh
| | - Jeffrey I. Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110 USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
4
|
Martín-Masot R, Jiménez-Muñoz M, Herrador-López M, Navas-López VM, Obis E, Jové M, Pamplona R, Nestares T. Metabolomic Profiling in Children with Celiac Disease: Beyond the Gluten-Free Diet. Nutrients 2023; 15:2871. [PMID: 37447198 DOI: 10.3390/nu15132871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Celiac disease (CD) is included in the group of complex or multifactorial diseases, i.e., those caused by the interaction of genetic and environmental factors. Despite a growing understanding of the pathophysiological mechanisms of the disease, diagnosis is still often delayed and there are no effective biomarkers for early diagnosis. The only current treatment, a gluten-free diet (GFD), can alleviate symptoms and restore intestinal villi, but its cellular effects remain poorly understood. To gain a comprehensive understanding of CD's progression, it is crucial to advance knowledge across various scientific disciplines and explore what transpires after disease onset. Metabolomics studies hold particular significance in unravelling the complexities of multifactorial and multisystemic disorders, where environmental factors play a significant role in disease manifestation and progression. By analyzing metabolites, we can gain insights into the reasons behind CD's occurrence, as well as better comprehend the impact of treatment initiation on patients. In this review, we present a collection of articles that showcase the latest breakthroughs in the field of metabolomics in pediatric CD, with the aim of trying to identify CD biomarkers for both early diagnosis and treatment monitoring. These advancements shed light on the potential of metabolomic analysis in enhancing our understanding of the disease and improving diagnostic and therapeutic strategies. More studies need to be designed to cover metabolic profiles in subjects at risk of developing the disease, as well as those analyzing biomarkers for follow-up treatment with a GFD.
Collapse
Affiliation(s)
- Rafael Martín-Masot
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
| | - María Jiménez-Muñoz
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Marta Herrador-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Víctor Manuel Navas-López
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Malaga, 29010 Málaga, Spain
| | - Elia Obis
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Mariona Jové
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - Teresa Nestares
- Institute of Nutrition and Food Technology "José MataixVerdú" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18071 Granada, Spain
- Department of Physiology, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| |
Collapse
|
5
|
Wei X, Tan X, Chen Q, Jiang Y, Wu G, Ma X, Fu J, Li Y, Gang K, Yang Q, Ni R, He J, Luo L. Extensive jejunal injury is repaired by migration and transdifferentiation of ileal enterocytes in zebrafish. Cell Rep 2023; 42:112660. [PMID: 37342912 DOI: 10.1016/j.celrep.2023.112660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/07/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
A major cause of intestinal failure (IF) is intestinal epithelium necrosis and massive loss of enterocytes, especially in the jejunum, the major intestinal segment in charge of nutrient absorption. However, mechanisms underlying jejunal epithelial regeneration after extensive loss of enterocytes remain elusive. Here, we apply a genetic ablation system to induce extensive damage to jejunal enterocytes in zebrafish, mimicking the jejunal epithelium necrosis that causes IF. In response to injury, proliferation and filopodia/lamellipodia drive anterior migration of the ileal enterocytes into the injured jejunum. The migrated fabp6+ ileal enterocytes transdifferentiate into fabp2+ jejunal enterocytes to fulfill the regeneration, consisting of dedifferentiation to precursor status followed by redifferentiation. The dedifferentiation is activated by the IL1β-NFκB axis, whose agonist promotes regeneration. Extensive jejunal epithelial damage is repaired by the migration and transdifferentiation of ileal enterocytes, revealing an intersegmental migration mechanism of intestinal regeneration and providing potential therapeutic targets for IF caused by jejunal epithelium necrosis.
Collapse
Affiliation(s)
- Xiangyong Wei
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Xinmiao Tan
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Qi Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Yan Jiang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Guozhen Wu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Xue Ma
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Jialong Fu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Yongyu Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Kai Gang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Qifen Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Rui Ni
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
6
|
Perez-Diaz-Del-Campo N, Castelnuovo G, Ribaldone DG, Caviglia GP. Fecal and Circulating Biomarkers for the Non-Invasive Assessment of Intestinal Permeability. Diagnostics (Basel) 2023; 13:diagnostics13111976. [PMID: 37296827 DOI: 10.3390/diagnostics13111976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
The study of intestinal permeability is gaining growing interest due to its relevance in the onset and progression of several gastrointestinal and non-gastrointestinal diseases. Though the involvement of impaired intestinal permeability in the pathophysiology of such diseases is recognized, there is currently a need to identify non-invasive biomarkers or tools that are able to accurately detect alterations in intestinal barrier integrity. On the one hand, promising results have been reported for novel in vivo methods based on paracellular probes, i.e., methods that can directly assess paracellular permeability and, on the other hand, on fecal and circulating biomarkers able to indirectly assess epithelial barrier integrity and functionality. In this review, we aimed to summarize the current knowledge on the intestinal barrier and epithelial transport pathways and to provide an overview of the methods already available or currently under investigation for the measurement of intestinal permeability.
Collapse
|
7
|
Pironi L. Definition, classification, and causes of short bowel syndrome. Nutr Clin Pract 2023; 38 Suppl 1:S9-S16. [PMID: 37115031 DOI: 10.1002/ncp.10955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 04/29/2023] Open
Abstract
The term "short bowel syndrome (SBS)" defines "the clinical feature associated with a remaining small bowel in continuity of less than 200 cm from the ligament of Treitz" and is characterized by malabsorption, diarrhea, fatty stools, malnutrition, and dehydration. SBS is the primary pathophysiological mechanism of chronic intestinal failure (CIF), defined as the "reduction of gut function below the minimum necessary for the absorption of macronutrients and/or water and electrolytes, such that intravenous supplementation (IVS) is required to maintain health and/or growth" in a metabolically stable patient. By contrast, the reduction of gut absorptive function that does not require IVS has been termed "intestinal insufficiency or deficiency" (II/ID). The classification of SBS can be categorized as follows: anatomical (anatomy and length of the residual bowel), evolutional (early, rehabilitative, and maintenance phases), pathophysiological (SBS with or without a colon in continuity), clinical (with II/ID or CIF), and severity of CIF (type and volume of the required IVS). Appropriate and homogeneous patient categorization is the mainstay of facilitating communication in clinical practice and in research.
Collapse
Affiliation(s)
- Loris Pironi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Centre for Chronic Intestinal Failure, IRCCS AOUBO, Bologna, Italy
| |
Collapse
|
8
|
Laterza L, Putignani L, Settanni CR, Petito V, Varca S, De Maio F, Macari G, Guarrasi V, Gremese E, Tolusso B, Wlderk G, Pirro MA, Fanali C, Scaldaferri F, Turchini L, Amatucci V, Sanguinetti M, Gasbarrini A. Ecology and Machine Learning-Based Classification Models of Gut Microbiota and Inflammatory Markers May Evaluate the Effects of Probiotic Supplementation in Patients Recently Recovered from COVID-19. Int J Mol Sci 2023; 24:ijms24076623. [PMID: 37047594 PMCID: PMC10094838 DOI: 10.3390/ijms24076623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Gut microbiota (GM) modulation can be investigated as possible solution to enhance recovery after COVID-19. An open-label, single-center, single-arm, pilot, interventional study was performed by enrolling twenty patients recently recovered from COVID-19 to investigate the role of a mixed probiotic, containing Lactobacilli, Bifidobacteria and Streptococcus thermophilus, on gastrointestinal symptoms, local and systemic inflammation, intestinal barrier integrity and GM profile. Gastrointestinal Symptom Rating Scale, cytokines, inflammatory, gut permeability, and integrity markers were evaluated before (T0) and after 8 weeks (T1) of probiotic supplementation. GM profiling was based on 16S-rRNA targeted-metagenomics and QIIME 2.0, LEfSe and PICRUSt computational algorithms. Multiple machine learning (ML) models were trained to classify GM at T0 and T1. A statistically significant reduction of IL-6 (p < 0.001), TNF-α (p < 0.001) and IL-12RA (p < 0.02), citrulline (p value < 0.001) was reported at T1. GM global distribution and microbial biomarkers strictly reflected probiotic composition, with a general increase in Bifidobacteria at T1. Twelve unique KEGG orthologs were associated only to T0, including tetracycline resistance cassettes. ML classified the GM at T1 with 100% score at phylum level. Bifidobacteriaceae and Bifidobacterium spp. inversely correlated to reduction of citrulline and inflammatory cytokines. Probiotic supplementation during post-COVID-19 may trigger anti-inflammatory effects though Bifidobacteria and related-metabolism enhancement.
Collapse
Affiliation(s)
- Lucrezia Laterza
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostics and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Immunology, Rheumatology and Infectious diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Carlo Romano Settanni
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Valentina Petito
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Simone Varca
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Flavio De Maio
- Laboratorio di Microbiologia Clinica, Dipartimento di Scienze di Laboratorio ed Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | | | | | - Elisa Gremese
- Immunology Facility, Gstep, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Barbara Tolusso
- Immunology Facility, Gstep, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Giulia Wlderk
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Maria Antonia Pirro
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Caterina Fanali
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Laura Turchini
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Valeria Amatucci
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Maurizio Sanguinetti
- Laboratorio di Microbiologia Clinica, Dipartimento di Scienze di Laboratorio ed Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- CeMAD, Digestive Disease Center, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| |
Collapse
|
9
|
Haroun E, Kumar PA, Saba L, Kassab J, Ghimire K, Dutta D, Lim SH. Intestinal barrier functions in hematologic and oncologic diseases. J Transl Med 2023; 21:233. [PMID: 37004099 PMCID: PMC10064590 DOI: 10.1186/s12967-023-04091-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023] Open
Abstract
The intestinal barrier is a complex structure that not only regulates the influx of luminal contents into the systemic circulation but is also involved in immune, microbial, and metabolic homeostasis. Evidence implicating disruption in intestinal barrier functions in the development of many systemic diseases, ranging from non-alcoholic steatohepatitis to autism, or systemic complications of intestinal disorders has increased rapidly in recent years, raising the possibility of the intestinal barrier as a potential target for therapeutic intervention to alter the course and mitigate the complications associated with these diseases. In addition to the disease process being associated with a breach in the intestinal barrier functions, patients with hematologic and oncologic diseases are particularly at high risks for the development of increased intestinal permeability, due to the frequent use of broad-spectrum antibiotics and chemoradiation. They also face a distinct challenge of being intermittently severely neutropenic due to treatment of the underlying conditions. In this review, we will discuss how hematologic and oncologic diseases are associated with disruption in the intestinal barrier and highlight the complications associated with an increase in the intestinal permeability. We will explore methods to modulate the complication. To provide a background for our discussion, we will first examine the structure and appraise the methods of evaluation of the intestinal barrier.
Collapse
Affiliation(s)
- Elio Haroun
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Prashanth Ashok Kumar
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Ludovic Saba
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Joseph Kassab
- Department of Medicine, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Krishna Ghimire
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA
| | - Dibyendu Dutta
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA.
| | - Seah H Lim
- Division of Hematology and Oncology, State University of New York Upstate Medical University, SUNY Upstate Medical University, 750 E Adams, Syracuse, NY, 13210, USA.
| |
Collapse
|
10
|
Mashaqi S, Rangan P, Saleh AA, Abraham I, Gozal D, Quan SF, Parthasarathy S. Biomarkers of gut barrier dysfunction in obstructive sleep apnea: A systematic review and meta-analysis. Sleep Med Rev 2023; 69:101774. [PMID: 37028145 DOI: 10.1016/j.smrv.2023.101774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023]
Abstract
We conducted this systematic review and meta-analysis to evaluate the impact of obstructive sleep apnea (OSA) on gut barrier dysfunction as represented by the following biomarkers: zonulin, lipopolysaccharide, lipopolysaccharide binding protein, intestinal fatty acid binding protein, and lactic acid. A comprehensive search of the literature was conducted in Ovid MEDLINE, Embase, Scopus, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov without language restrictions from inception to October 2022. The analysis of all outcomes was performed using a random-effects model. We included eight studies (seven cross sectional and one case control) in the final quantitative synthesis with a total of 897 patients. We concluded that OSA was associated with higher levels of gut barrier dysfunction biomarkers [Hedges' g = 0.73 (95%CI 0.37-1.09, p < 0.01). Biomarker levels were positively correlated with the apnea-hypopnea index [r = 0.48 (95%CI 0.35-0.6, p < 0.01)] and oxygen desaturation index [r = 0.30 (95%CI 0.17-0.42, p < 0.01)], and negatively correlated with the nadir oxygen desaturation values [r = -0.45 (95%CI - 0.55 - - 0.32, p < 0.01). Our systematic review and meta-analysis suggests that OSA is associated with gut barrier dysfunction. Furthermore, OSA severity appears to be correlated with higher biomarkers of gut barrier dysfunction. PROSPERO REGISTRATION NUMBER: CRD42022333078.
Collapse
Affiliation(s)
- Saif Mashaqi
- Department of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The University of Arizona, College of Medicine Tucson, 1625 N Campbell Ave, Tucson, AZ, 85719, USA.
| | - Pooja Rangan
- Division of Clinical Data Analytics and Decision Support, Department of Internal Medicine, The University of Arizona College of Medicine Phoenix, AZ, USA.
| | - Ahlam A Saleh
- Health Sciences Library, The University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85724, USA.
| | - Ivo Abraham
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA; Department of Family and Community Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA.
| | - David Gozal
- Department of Child Health, University of Missouri, Columbia, MO, USA.
| | - Stuart F Quan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Harvard Medical School, USA.
| | - Sairam Parthasarathy
- Department of Pulmonary, Allergy, Critical Care, and Sleep Medicine, The University of Arizona, College of Medicine Tucson, USA.
| |
Collapse
|
11
|
Onuk S, Ozer NT, Ozel M, Sipahioglu H, Kahriman G, Baskol G, Temel S, Gundogan K, Akin A. Gastric ultrasound, citrulline, and intestinal fatty acid-binding protein as markers of gastrointestinal dysfunction in critically ill patients: A pilot prospective cohort study. JPEN J Parenter Enteral Nutr 2023; 47:429-436. [PMID: 36609803 DOI: 10.1002/jpen.2473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Gastrointestinal (GI) dysfunction is common in the intensive care unit (ICU), although there is no consensus on biomarkers of GI dysfunction. We aimed to evaluate ultrasound-based gastric antrum measurements and serum intestinal fatty acid-binding protein (IFABP) and citrulline levels in relation to GI dysfunction in critically ill patients. METHODS Adult critically ill patients receiving enteral nutrition and stayed for in the ICU for ≥48 h was included. GI dysfunction was described using Gastrointestinal Dysfunction Score (GIDS). Gastric antrum measurements, including craniocaudal (CC) diameter, anteroposterior diameter, and antral-cross sectional area (CSA), as well as serum levels for IFABP and citrulline, were prospectively recorded at baseline and on day 3 and day 5 of enteral nutrition. The receiver operating characteristic (ROC) analysis was performed to evaluate gastric ultrasound parameters, serum IFABP, and citrulline concentrations in predicting GI dysfunction. RESULTS Thirty-nine participants with a median age of 60 years were recruited and 46.2% of participants had GI dysfunction. ROC analysis revealed that the cutoff value of CSA score to predict GI dysfunction was 4.48 cm2 , which provided 72.7% sensitivity and 77.2% specificity (area under the curve = 0.768, 95% CI: 0.555-0.980). At baseline, gastric residual volume was highly correlated with CC diameter and CSA (r = 0.764, P < 0.001 and r = 0.675, P < 0.001, respectively). Serum IFABP and citrulline levels had no correlation with GI dysfunction or gastric ultrasound parameters (P > 0.05). CONCLUSION CSA was associated with GI dysfunction in critically ill patients. Serum IFABP and citrulline concentrations were poor in predicting GI dysfunction.
Collapse
Affiliation(s)
- Sevda Onuk
- Division of Intensive Care Unit, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Nurhayat Tugra Ozer
- Department of Clinical Nutrition, Health Science Institute, Erciyes University, Kayseri, Turkey
| | - Merve Ozel
- Department of Clinical Biochemistry, Erciyes University, Kayseri, Turkey
| | - Hilal Sipahioglu
- Division of Intensive Care Unit, Kayseri City Training and Research Hospital, Kayseri, Turkey
| | - Güven Kahriman
- Department of Radiology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Gulden Baskol
- Department of Clinical Biochemistry, Erciyes University, Kayseri, Turkey
| | - Sahin Temel
- Division of Intensive Care, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Kursat Gundogan
- Division of Intensive Care, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Aynur Akin
- Division of Anestesiology and Reanimation Intensive Care, School of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
12
|
Jardi F, Kelly C, Teague C, Fowler-Williams H, Sevin DC, Rodrigues D, Jo H, Ferreira S, Herpers B, Van Heerden M, de Kok T, Pin C, Lynch A, Duckworth CA, De Jonghe S, Lammens L, Pritchard DM. Mouse organoids as an in vitro tool to study the in vivo intestinal response to cytotoxicants. Arch Toxicol 2023; 97:235-54. [PMID: 36203040 DOI: 10.1007/s00204-022-03374-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/05/2022] [Indexed: 01/19/2023]
Abstract
Cross-species comparison of drug responses at the organoid level could help to determine the human relevance of findings from animal studies. To this end, we first need to evaluate the in vitro to in vivo translatability of preclinical organoids. Here, we used 5-fluorouracil (5-FU) as an exemplar drug to test whether the in vivo gut response to this cytotoxicant was preserved in murine intestinal organoids. Mice treated with 5-FU at 20 or 50 mg/kg IV (low and high dose, respectively) displayed diarrhea at clinically relevant exposures. 5-FU also induced intestinal lesions, increased epithelial apoptosis, and decreased proliferation in a dose-dependent manner. To enable comparison between the in vitro and in vivo response, top nominal in vitro drug concentrations that caused significant cytotoxicity were chosen (dose range 1-1000 µM). The inferred intracellular concentration in organoids at 1000 µM was within the tissue exposure range related to intestinal toxicity in vivo. 5-FU at ≥ 100 µM decreased ATP levels and increased Caspase-3 activity in intestinal organoids. In keeping with the in vivo findings, 5-FU increased the percentage of Caspase-3-positive cells and reduced Ki67 staining. At the transcriptome level, there was an overlap in the activity of pathways related to 5-FU's mode of action, lipid and cholesterol metabolism and integrin signaling across in vivo gut and organoids. The predicted activity state of upstream regulators was generally well preserved between setups. Collectively, our results suggest that despite their inherent limitations, organoids represent an adequate tool to explore the intestinal response to cytotoxicants.
Collapse
|
13
|
DIK I, HATIPOGLU D, GULERSOY E. Comparison of some cytokines, acute phase proteins and citrulline levels in healthy and canine distemper infected dogs. J Vet Med Sci 2023; 85:76-82. [PMID: 36418074 PMCID: PMC9887225 DOI: 10.1292/jvms.22-0281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Canine distemper virus (CDV) is the etiological agent of severe disease in domestic and wild carnivores. Clinical diagnosis of CDV is challenging because of its similarity to other canine respiratory and intestinal diseases. We aimed to determine certain cytokine (interleukin [IL]-1β, IL-2, IL-4, IL-6, IL-10, and tumor necrosis factor-α [TNF-α]), interferon (IFN)-γ, canine serum amyloid A (SAA), and canine citrulline (CIT) levels for the first time in CDV-positive dogs. For this purpose, 10 CDV-positive dogs with compatible clinical findings (i.e., neurological symptoms such as tremors and myoclonus, ocular and nasal discharge, and wheezing) and 10 healthy dogs based on the clinical examinations and rapid test results were enrolled. It was observed that the CIT, INF-γ, IL-1β, IL-2, IL-6, and TNF-α levels were significantly decreased in the CDV-positive dogs than that of the healthy ones (P<0.05). As a result, it was observed that CDV causes immunosuppression and accordingly, the inflammatory response might cause decreased cytokine and acute-phase protein synthesis. Therefore, it was concluded that further investigation of inflammatory pathways and CIT interactions may provide crucial clinical information at different stages of CDV, and aforementioned parameters may serve as important biomarkers for CDV in terms of demonstrating the presence of immunosuppression.
Collapse
Affiliation(s)
- Irmak DIK
- Department of Virology, Faculty of Veterinary Medicine, University of Selcuk, Konya, Türkiye
| | - Durmus HATIPOGLU
- Department of Physiology, Faculty of Veterinary Medicine, University of Selcuk, Konya, Türkiye,Correspondence to: Hatıpoglu D: , Department of Virology, Faculty of Veterinary Medicine, University of Selcuk, 42075, Konya, Türkiye
| | - Erdem GULERSOY
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Harran, Şanlıurfa, Türkiye
| |
Collapse
|
14
|
Suwanphoerung W, Klinmalai C, Rattanasiri S, Pakakasama S, Anurathapan U, Hongeng S, Chongviriyaphan N, Apiwattanakul N. Association of zinc deficiency with infectious complications in pediatric hematopoietic stem cell transplantation patients. PLoS One 2022; 17:e0279439. [PMID: 36574381 PMCID: PMC9794056 DOI: 10.1371/journal.pone.0279439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Zinc plays essential roles in immune function and epithelial integrity. Patients undergoing hematopoietic stem cell transplantation (HSCT) often have low plasma zinc levels because of poor intake and diarrhea. We hypothesized that patients with zinc deficiency before HSCT had worse infectious complications after HSCT compared with patients with normal zinc levels. Citrulline, a marker of intestinal integrity, was also hypothesized to be lower in patients with zinc deficiency. PATIENTS AND METHODS Thirty patients undergoing HSCT at Ramathibodi Hospital during March 2020-September 2021 were enrolled. Blood samples for plasma zinc and citrulline were collected during the HSCT period. The 14- and 90-day outcomes after HSCT were prospectively recorded. RESULTS Twelve of 30 (40%) patients had zinc deficiency before HSCT. Zinc-deficient patients were younger (median (interquartile range): 6 (8.8) vs 13 (5.8) years old; p = 0.017). Zinc levels tended to increase after admission in both groups. Patients with zinc deficiency had lower citrulline levels than those with normal zinc levels. Citrulline levels decreased in both groups after stem cell infusion, and the level was not significantly different between the two groups. Zinc-deficient patients had a higher rate of bacterial infection within 90 days after HSCT than those with normal zinc levels (6 in 12 patients (50.0%) vs 1 in 18 patients (5.6%); odds ratio [OR]: 17.0; 95% confidence interval [CI]: 1.68-171.70; p = 0.016). This remained significant after adjustments for age (adjusted OR: 12.31; 95% CI: 1.084-139.92; p = 0.043). CONCLUSION The prevalence of zinc deficiency in pediatric patients undergoing HSCT was high. Zinc-deficient patients had lower citrulline levels and higher incidence of bacterial infection after HSCT. However, citrulline level was not different between patients with and without bacterial infections. It is worth to investigate whether zinc supplementation before HSCT can reduce bacterial infection after HSCT.
Collapse
Affiliation(s)
- Warangkhana Suwanphoerung
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chompunuch Klinmalai
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasivimol Rattanasiri
- Section for Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Samart Pakakasama
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nalinee Chongviriyaphan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nopporn Apiwattanakul
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
15
|
Stoeckle SD, Timmermann D, Merle R, Gehlen H. Plasma Amino Acid Concentration in Obese Horses with/without Insulin Dysregulation and Laminitis. Animals (Basel) 2022; 12. [PMID: 36552500 DOI: 10.3390/ani12243580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Laminitic horses commonly suffer from an endocrine disease such as equine metabolic syndrome. Hyperinsulinemia is considered a key factor in the pathogenesis of laminitis. Since insulin also affects protein turnover in the body, the resting plasma amino acid concentrations of obese horses that were presented for a combined glucose insulin test (CGIT) were determined. In total, 25 obese horses and two lean horses with recurrent laminitis underwent a CGIT. Of these, five were not insulin dysregulated (obese), 14 were insulin dysregulated (ID), and eight were insulin-dysregulated and laminitic (IDL). Significant differences in the resting concentrations between obese and insulin dysregulated and laminitic (citrulline p = 0.038, obese: 73.001 ± 12.661 nmol/mL, IDL: 49.194 ± 15.486 nmol/mL; GABA p = 0.02, obese: 28.234 ± 3.885 nmol/mL, IDL: 16.697 ± 1.679 nmol/mL; methionine p = 0.018, obese: 28.691 ± 5.913 nmol/mL, IDL: 20.143 ± 3.09 nmol/mL) as well as between insulin dysregulated individuals with and without laminitis (GABA p < 0.001, ID: 28.169 ± 6.739 nmol/mL) regarding three amino acids were determined. This may be an interesting approach, especially for diagnostic testing and possibly also for the feed supplements of horses at risk of developing laminitis. However, further research, including a higher number of cases, is required.
Collapse
|
16
|
Møller DL, Kielsen K, Nielsen CH, Sengeløv H, Pedersen AE, Ryder LP, Müller K. Thymic stromal lymphopoietin levels after allogeneic hematopoietic stem cell transplantation. Immunopharmacol Immunotoxicol 2022; 44:1004-1012. [PMID: 35899395 DOI: 10.1080/08923973.2022.2102989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. METHODS We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry. RESULTS TSLP levels rose after initiation of the conditioning to peak at day +21 after HSCT (p = .03), where TSLP levels correlated with counts of neutrophils (rho = 0.36, p = .04), monocytes (rho = 0.58, p = .006), and lymphocytes (rho = 0.59, p = .02). Overall absolute TSLP levels were not associated with acute or chronic graft-vs-host disease (a/cGvHD). However, patients mounting a sustained increase in TSLP levels at day +90 had a higher risk of cGvHD compared to patients who had returned to pre-conditioning levels at that stage (cumulative incidence: 77% vs. 38%, p = .01). CONCLUSION In conclusion, this study suggests a role of TSLP in immune reconstitution and alloreactivity post-HSCT. lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry.
Collapse
Affiliation(s)
- Dina Leth Møller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Katrine Kielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henrik Sengeløv
- Department of Hematology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Lars Peter Ryder
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Stoeckle SD, Timmermann D, Merle R, Gehlen H. Plasma Amino Acids in Horses Suffering from Pituitary Pars Intermedia Dysfunction. Animals (Basel) 2022; 12. [PMID: 36496836 DOI: 10.3390/ani12233315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Pituitary pars intermedia dysfunction is one of the most common diseases of aged horses and ponies. In Parkinson's disease, which is, similar to PPID, a disease that involves oxidative damage to dopaminergic pathways but with different clinical signs, alterations to the serum amino acid profile have been reported. To examine changes in the plasma amino acid profile in horses with PPID, EDTA plasma of horses that were presented for various reasons that required laboratory examinations of blood anticoagulated with EDTA was collected. With this plasma, the basal ACTH concentration as well as the amino acid profile was determined. Horses were considered PPID patients if the ACTH concentration was ≥ 100 pg/mL, i.e., they would be considered affected at any time. Horses were defined as non-PPID (nPPID) patients if the ACTH concentration was below 30 pg/mL. Horses receiving pergolide with ACTH ≤ 30 pg/mL were allocated to the group PPIDrr (PPID, ACTH in reference range) and horses receiving pergolide with ACTH ≥ 100 pg/mL to the group PPIDarr (PPID, ACTH above reference range). In total, 93 horses were examined, including 88 horses at the clinic and 5 horses at a private practice. Of these, 53 horses fulfilled the inclusion criteria (ACTH ≤ 30 pg/mL or ACTH ≥ 100 pg/mL). A total of 25 horses were diagnosed as nPPID, 20 as PPID, 5 as PPIDrr, and 3 as PPIDarr. Arginine was significantly higher in PPIDrr than in PPID and nPPID, asparagine was significantly higher in PPID, PPIDrr, and PPIDarr than in nPPID, citrulline was significantly higher in PPIDrr than in nPPID and PPID, cysteine was significantly lower in PPIDrr than in PPID, nPPID, and PPIDarr, and glutamine was significantly higher in PPID and PPIDarr than in nPPID. Especially, asparagine, citrulline, and glutamine may be potential diagnostic markers and may offer interesting approaches for research regarding amino supplementation in PPID.
Collapse
|
18
|
Zhang H, Zheng Y, Zha X, Ma Y, Liu X, Elsabagh M, Wang H, Wang M. Dietary L-Arginine or N-Carbamylglutamate Alleviates Colonic Barrier Injury, Oxidative Stress, and Inflammation by Modulation of Intestinal Microbiota in Intrauterine Growth-Retarded Suckling Lambs. Antioxidants (Basel) 2022; 11:antiox11112251. [PMID: 36421439 PMCID: PMC9687183 DOI: 10.3390/antiox11112251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
Our previous studies have revealed that dietary N-carbamylglutamate (NCG) and L-arginine (Arg) supplementation improves redox status and suppresses apoptosis in the colon of suckling Hu lambs with intrauterine growth retardation (IUGR). However, no studies have reported the function of Arg or NCG in the colonic microbial communities, barrier function, and inflammation in IUGR-suckling lambs. This work aimed to further investigate how dietary Arg or NCG influences the microbiota, barrier function, and inflammation in the colon of IUGR lambs. Forty-eight newborn Hu lambs of 7 d old were assigned to four treatment groups (n = 12 per group; six male, six female) as follows: CON (normal birth weight, 4.25 ± 0.14 kg), IUGR (3.01 ± 0.12 kg), IUGR + Arg (2.99 ± 0.13 kg), and IUGR + NCG (3.03 ± 0.11 kg). A total of 1% Arg or 0.1% NCG was supplemented in a basal diet of milk replacer, respectively. Lambs were fed the milk replacer for 21 d until 28 d after birth. Compared to the non-supplemented IUGR lambs, the transepithelial electrical resistance (TER) was higher, while fluorescein isothiocyanate dextran 4 kDa (FD4) was lower in the colon of the NCG- or Arg-supplemented IUGR lambs (p < 0.05). The IUGR lambs exhibited higher (p < 0.05) colonic interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, reactive oxygen species (ROS), and malondialdehyde (MDA) levels than the CON lambs; the detrimental effects of IUGR on colonic proinflammatory cytokine concentrations and redox status were counteracted by dietary Arg or NCG supplementation. Both IUGR + Arg and IUGR + NCG lambs exhibited an elevated protein and mRNA expression of Occludin, Claudin-1, and zonula occludens-1 (ZO-1) compared to the IUGR lambs (p < 0.05). Additionally, the lipopolysaccharide (LPS) concentration was decreased while the levels of acetate, butyrate, and propionate were increased in IUGR + Arg and IUGR + NCG lambs compared to the IUGR lambs (p < 0.05). The relative abundance of Clostridium, Lactobacillus, and Streptococcus was lower in the colonic mucosa of the IUGR lambs than in the CON lambs (p < 0.05) but was restored upon the dietary supplementation of Arg or NCG to the IUGR lambs (p < 0.05). Both Arg and NCG can alleviate colonic barrier injury, oxidative stress (OS), and inflammation by the modulation of colonic microbiota in IUGR-suckling lambs. This work contributes to improving knowledge about the crosstalk among gut microbiota, immunity, OS, and barrier function and emphasizes the potential of Arg or NCG in health enhancement as feed additives in the early life nutrition of ruminants.
Collapse
Affiliation(s)
- Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yi Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyun Liu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde 51240, Turkey
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.W.); (M.W.)
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.W.); (M.W.)
| |
Collapse
|
19
|
Soares I, Belote BL, Santin E, Dal Pont GC, Kogut MH. Morphological Assessment and Biomarkers of Low-Grade, Chronic Intestinal Inflammation in Production Animals. Animals (Basel) 2022; 12:3036. [PMID: 36359160 PMCID: PMC9654368 DOI: 10.3390/ani12213036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Production animals are continuously exposed to environmental and dietary factors that might induce a state of low-grade, chronic intestinal inflammation. This condition compromises the productive performance and well-fare of these animals, requiring studies to understand what causes it and to develop control strategies. An intestinal inflammatory process is generally associated with alterations in the structure and functionality of its wall, resulting in the release of cellular components into the blood and/or feces. These components can act as biomarkers, i.e., they are measured to identify and quantify an inflammatory process without requiring invasive methods. In this review we discuss the mechanisms of low-grade inflammation, its effects on animal production and sustainability, and the identification of biomarkers that could provide early diagnosis of this process and support studies of useful interventional strategies. Abstract The complex interaction between the intestinal mucosa, the gut microbiota, and the diet balances the host physiological homeostasis and is fundamental for the maximal genetic potential of production animals. However, factors such as chemical and physical characteristics of the diet and/or environmental stressors can continuously affect this balance, potentially inducing a state of chronic low-grade inflammation in the gut, where inflammatory parameters are present and demanding energy, but not in enough intensity to provoke clinical manifestations. It’s vital to expand the understanding of inflammation dynamics and of how they compromise the function activity and microscopic morphology of the intestinal mucosa. These morphometric alterations are associated with the release of structural and functional cellular components into the feces and the blood stream creating measurable biomarkers to track this condition. Moreover, the identification of novel, immunometabolic biomarkers can provide dynamic and predictors of low-grade chronic inflammation, but also provide indicators of successful nutritional or feed additive intervention strategies. The objective of this paper is to review the mechanisms of low-grade inflammation, its effects on animal production and sustainability, and the biomarkers that could provide early diagnosis of this process and support studies of useful interventional strategies.
Collapse
|
20
|
Li Q, Zhou HB, Liu JQ, Bai WF, Wang J, Yang ZJ, Qiu M, Chang H, Shi SL. The intervention effect of Amygdalus mongolica oil on the metabolomics and intestinal flora in pulmonary fibrosis. Front Pharmacol 2022; 13:1037563. [PMID: 36386194 PMCID: PMC9663812 DOI: 10.3389/fphar.2022.1037563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/14/2022] [Indexed: 11/28/2022] Open
Abstract
Amygdalus mongolica oil is rich in unsaturated fatty acids such as inoleic acid (47.11%) and oleic acid (23.81%). Our research demonstrates that it exerts a protective effect on rat models of pulmonary fibrosis, however, little is known regarding the underlying mechanism of action. This study aimed to characterize the therapeutic mechanism of action of A. mongolica oil on bleomycin-induced pulmonary fibrosis in rats. A. mongolica oil appears to regulate the levels of potential key serum biomarkers which include tetrahydrobiopterin, L-serine, citrulline and estradiol to participate in folate biosynthesis, glycine, serine and threonine metabolism, arginine biosynthesis and steroid hormone biosynthesis. And it also enriched intestinal microbial abundance, homogeneity and modulated the abundance of Duncaniell, Desulfovibrio, Peptococcaceae_unclassified, Dubosiella, Tyzzerella, Lachnospiraceae_NK4A136_group, Lactobacillus, Clostridiales_unclassified to exert a protective effect against pulmonary fibrosis. A. mongolica oil appears to confer protective effects against pulmonary fibrosis by affecting the level of pulmonary fibrosis metabolites and the abundance of related intestinal flora through multiple targets, as evidenced by our untargeted LC-MS/MS metabonomics evaluation and 16S rDNA sequencing technology.
Collapse
Affiliation(s)
- Qian Li
- 1Department of Pharmacy, Baotou Medical College, Baotou, China,2Inner Mongolia Maternal and Child Health Care Hospital, Hohhot, China
| | - Hong-Bing Zhou
- 1Department of Pharmacy, Baotou Medical College, Baotou, China,3Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, China
| | - Jia-Qi Liu
- 1Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Wan-Fu Bai
- 1Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Jia Wang
- 1Department of Pharmacy, Baotou Medical College, Baotou, China
| | - Zhan-Jun Yang
- 3Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, China
| | - Min Qiu
- 1Department of Pharmacy, Baotou Medical College, Baotou, China,3Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, China
| | - Hong Chang
- 1Department of Pharmacy, Baotou Medical College, Baotou, China,*Correspondence: Hong Chang, ; Song-Li Shi,
| | - Song-Li Shi
- 1Department of Pharmacy, Baotou Medical College, Baotou, China,3Institute of Bioactive Substance and Function of Mongolian Medicine and Chinese Materia Medica, Baotou Medical College, Baotou, China,*Correspondence: Hong Chang, ; Song-Li Shi,
| |
Collapse
|
21
|
Garg S, Garg TK, Miousse IR, Wise SY, Fatanmi OO, Savenka AV, Basnakian AG, Singh VK, Hauer-jensen M. Effects of Gamma-Tocotrienol on Partial-Body Irradiation-Induced Intestinal Injury in a Nonhuman Primate Model. Antioxidants (Basel) 2022; 11:1895. [PMID: 36290618 PMCID: PMC9598988 DOI: 10.3390/antiox11101895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Exposure to high doses of radiation, accidental or therapeutic, often results in gastrointestinal (GI) injury. To date, there are no therapies available to mitigate GI injury after radiation exposure. Gamma-tocotrienol (GT3) is a promising radioprotector under investigation in nonhuman primates (NHP). We have shown that GT3 has radioprotective function in intestinal epithelial and crypt cells in NHPs exposed to 12 Gy total-body irradiation (TBI). Here, we determined GT3 potential in accelerating the GI recovery in partial-body irradiated (PBI) NHPs using X-rays, sparing 5% bone marrow. Sixteen rhesus macaques were treated with either vehicle or GT3 24 h prior to 12 Gy PBI. Structural injuries and crypt survival were examined in proximal jejunum on days 4 and 7. Plasma citrulline was assessed using liquid chromatography–tandem mass spectrometry (LC-MS/MS). Crypt cell proliferation and apoptotic cell death were evaluated using Ki-67 and TUNEL staining. PBI significantly decreased mucosal surface area and reduced villous height. Interestingly, GT3 increased crypt survival and enhanced stem cell proliferation at day 4; however, the effects seemed to be minimized by day 7. GT3 did not ameliorate a radiation-induced decrease in citrulline levels. These data suggest that X-rays induce severe intestinal injury post-PBI and that GT3 has minimal radioprotective effect in this novel model.
Collapse
|
22
|
Abstract
Metabolic rewiring is a characteristic hallmark of cancer cells. This phenomenon sustains uncontrolled proliferation and resistance to apoptosis by increasing nutrients and energy supply. However, reprogramming comes together with vulnerabilities that can be used against tumor and can be applied in targeted therapy. In the last years, the genetic background of tumors has been identified thoroughly and new therapies targeting those mutations tested. Nevertheless, we propose that targeting the phenotype of cancer cells could be another way of treatment aiming to avoid drug resistance and non-responsiveness of cancer patients. Amino acid metabolism is part of the altered processes in cancer cells. Amino acids are building blocks and also sensors of signaling pathways regulating main biological processes. In this comprehensive review, we described four amino acids (asparagine, arginine, methionine, and cysteine) which have been actively investigated as potential targets for anti-tumor therapy. Asparagine depletion is successfully used for decades in the treatment of acute lymphoblastic leukemia and there is a strong implication to apply it to other types of tumors. Arginine auxotrophic tumors are great candidates for arginine-starvation therapy. Higher requirement for essential amino acids such as methionine and cysteine point out promising targetable weaknesses of cancer cells.
Collapse
Affiliation(s)
- Lucie Safrhansova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Dept. of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Hlozkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Dept. of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Julia Starkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic; Dept. of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic; University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
23
|
Dai D, Dai F, Chen J, Jin M, Li M, Hu D, Liu Z, Zhang Z, Xu F, Chen WH. Integrated multi-omics reveal important roles of gut contents in intestinal ischemia–reperfusion induced injuries in rats. Commun Biol 2022; 5:938. [PMID: 36085351 PMCID: PMC9463172 DOI: 10.1038/s42003-022-03887-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/08/2022] [Indexed: 12/13/2022] Open
Abstract
Intestinal ischemia–reperfusion (IIR) is a life-threatening clinical event with damaging signals whose origin and contents are unclear. Here we observe that IIR significantly affect the metabolic profiles of most organs by unbiased organ-wide metabolic analysis of gut contents, blood, and fifteen organs in rats (n = 29). Remarkably, correlations between gut content metabolic profiles and those of other organs are the most significant. Gut contents are also the only ones to show dynamic correlations during IIR. Additionally, according to targeted metabolomics analysis, several neurotransmitters are considerably altered in the gut during IIR, and displayed noteworthy correlations with remote organs. Likewise, metagenomics analysis (n = 35) confirm the effects of IIR on gut microbiota, and identify key species fundamental to the changes in gut metabolites, particularly neurotransmitters. Our multi-omics results establish key roles of gut contents in IIR induced remote injury and provide clues for future exploration. Die Dai et al. evaluate changes in the metabolomic and gut microbiome in response to experimental intestinal ischemia reperfusion (IIR) injury in rats. Their results provide further insight into how gut contents contribute to widespread injury in IIR patients.
Collapse
|
24
|
Demiryas S, Orhan A. Advancements in Gastrointestinal System Biomarkers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The requirement for diagnostic surgical operations of gastrointestinal system
diseases significantly decreases with the help of proper diagnostic tools. These
modalities are also beneficial for identifying postoperative complications, allowing us
to diagnose them in earlier stages, and increasing the postoperative survival rates.
Biomarkers are considered an integral part of diagnostic examinations. Developments
of biomarkers used for diagnosing and treating abdominal diseases are essential for
improving our capabilities in non-invasive monitoring. In this chapter, we review both
the novel and the routine biomarkers in the diagnosis and follow-up of gastrointestinal
system diseases.
Collapse
Affiliation(s)
- Suleyman Demiryas
- Department of General Surgery, Cerrahpaşa School of Medicine, IIstanbul University –
Cerrahpaşa, Istanbul, Turkey
| | - Anıl Orhan
- Department of General Surgery, Haseki Training and Research Hospital, University of Health
Sciences, Istanbul, Turkey
| |
Collapse
|
25
|
van Rhee KP, de Vroom SL, van Hest RM, van der Linden PD, Tonino SH, Molendijk E, Mathôt RAA, Blijlevens NMA, Knibbe CAJ, Bruggemann RJM, Geerlings SE. Impact of mucositis on oral bioavailability and systemic exposure of ciprofloxacin Gram-negative infection prophylaxis in patients with haematological malignancies. J Antimicrob Chemother 2022; 77:3069-3076. [PMID: 35996887 DOI: 10.1093/jac/dkac283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [What about the content of this article? (0)] [Affiliation(s)] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/12/2022] Open
|