1
|
Kong F, Wang S, Zhang Y, Li C, Dai D, Guo C, Wang Y, Cao Z, Yang H, Bi Y, Wang W, Li S. Rumen microbiome associates with postpartum ketosis development in dairy cows: a prospective nested case-control study. MICROBIOME 2025; 13:69. [PMID: 40057813 PMCID: PMC11889851 DOI: 10.1186/s40168-025-02072-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Approximately, one-third of dairy cows suffer from postpartum diseases. Ketosis is considered an important inducer of other postpartum diseases by disrupting energy metabolism. Although the rumen microbiome may be involved in the etiology of ketosis by supplying volatile fatty acids, the rumen environmental dynamics of ketosis cows are unclear. Using multi-omics, this study aimed to elucidate changes in the rumen microbiome during parturition of ketosis cows and the association between the rumen microbiome and host energy metabolism. The study included 810 rumen content samples and 789 serum samples from day - 21 and 21 relative to calving day from 61 ketosis cows and 84 healthy cows. RESULTS In ketosis cows, the rumen bacterial composition after parturition changed dramatically and needed a longer time to restore. The molar proportions of propionate were lower in ketosis cows than those in healthy cows on days 3 and 7 and negatively correlated with the serum β-hydroxybutyrate (BHBA) levels. The fermentation sub-pathway of propionate metabolism and partial glucogenic amino acid pathways were downregulated on day 3. Prevotella, UBA1066, and microbiota diversity indices regulate serum BHBA and glucose (GLU) levels via arginine, alanine, glycine, or propionate. Propionate administration to ketosis cows potentially decreased the serum BHBA concentration. CONCLUSIONS Collectively, we found rumen disruption happened after calving among ketosis cows, and insufficient glycogenic substrates, such as propionate, may be related to ketosis development. The study findings have implications for the relationship between rumen microbiome dynamics and host energy metabolism, which lays the foundation for the future rumen microbiome investigation for improving postpartum management in cows. Video Abstract.
Collapse
Affiliation(s)
- Fanlin Kong
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shuo Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yijia Zhang
- Laboratory of Animal Neurobiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chen Li
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Dongwen Dai
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Cheng Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanliang Bi
- Key Laboratory for Dairy Cow Nutrition, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
2
|
Palmer AC, Hossain MI, Ali H, Ayesha K, Shaikh S, Islam MT, Johura FT, Pasqualino MM, Rahman H, Haque R, Alland K, Wu LSF, Schulze KJ, Chakraborty S, West KP, Alam M, Ahmed T, Labrique AB. Protein supplementation delivered alone or in combination with presumptive azithromycin treatment for enteric pathogens did not improve linear growth in Bangladeshi infants: results of a cluster-randomized controlled trial. Am J Clin Nutr 2025; 121:597-609. [PMID: 39788294 DOI: 10.1016/j.ajcnut.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Protein requirements established for healthy populations may be insufficient to support healthy growth in infants consuming largely cereal-based complementary foods and frequently exposed to enteric pathogens. OBJECTIVES This study aimed to assess independent and combined effects of protein supplementation and antibiotic treatment on linear growth of infants aged 6-12 mo. METHODS We conducted a 2 × 4 factorial cluster-randomized trial in northwestern Bangladesh, allocating 566 clusters to masked azithromycin (10 mg/kg × 3 d) or placebo at 6 and 9 mo of age and unmasked delivery of an egg white protein-rich blended food supplement (250 kcal; 10 g added protein), a rice-based isocaloric supplement, egg, or nutrition education from 6 to 12 mo. We measured length at 6 and 12 mo. For this cluster-level intention-to-treat analysis of the 2 × 2 antibiotic and protein interventions, we used multiple linear or log-binomial regression with generalized estimating equations to assess changes in length-for-age z (LAZ) score and stunting (LAZ < -2), respectively. RESULTS We enrolled 2055 infants (283 clusters) and included 1821 infants (281 clusters) with complete anthropometry data at 6 and 12 mo in our analysis. There were no significant interactions between the protein and antibiotic interventions for any outcomes. Independently, protein supplement did not improve LAZ (β: 0.05; 95% CI: 0.00, 0.11; P = 0.07) or reduce stunting (prevalence ratio: 1.12; 95% CI: 0.85, 1.49; P = 0.41) compared with the isocaloric supplement. The antibiotic intervention had no effect on LAZ (β: -0.05; 95% CI: -0.11, 0.01; P = 0.09) or stunting (prevalence ratio: 0.99; 95% CI: 0.75, 1.31; P = 0.96), relative to the placebo. CONCLUSIONS Supplementation to increase intakes of high-quality protein, provided with or without presumptive treatment for enteric pathogens, did not improve linear growth from 6 to 12 mo of age. This trial was registered at clinicaltrials.gov as NCT03683667.
Collapse
Affiliation(s)
- Amanda C Palmer
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.
| | | | - Hasmot Ali
- The JiVitA Project, Gaibandha, Bangladesh
| | | | | | | | | | - Monica M Pasqualino
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | | | - Kelsey Alland
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lee Shu-Fune Wu
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kerry J Schulze
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Keith P West
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | | | | | - Alain B Labrique
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
3
|
Zhang JY, Greenwald MJ, Rodriguez SH. Gut Microbiome and Retinopathy of Prematurity. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1683-1690. [PMID: 36780985 DOI: 10.1016/j.ajpath.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/13/2023]
Abstract
Retinopathy of prematurity (ROP), a leading cause of childhood blindness worldwide, is strongly associated with gestational age and weight at birth. Yet, many extremely preterm infants never develop ROP or develop only mild ROP with spontaneous regression. In addition, a myriad of other factors play a role in the retinal pathology, one of which may include the early gut microbiome. The complications associated with early gestational age include dysbiosis of the dynamic neonatal gut microbiome, as evidenced by the development of often concomitant conditions, such as necrotizing enterocolitis. Given this, alongside growing evidence for a gut-retina axis, there is an increasing interest in how the early intestinal environment may play a role in the pathophysiology of ROP. Potential mechanisms include dysregulation of vascular endothelial growth factor and insulin-like growth factor 1. Furthermore, the gut microbiome may be impacted by other known risk factors for ROP, such as intermittent hypoxia and sepsis treated with antibiotics. This mini-review summarizes the literature supporting these proposed avenues, establishing a foundation to guide future studies.
Collapse
Affiliation(s)
- Jason Y Zhang
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois; Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Mark J Greenwald
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Sarah H Rodriguez
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois.
| |
Collapse
|
4
|
Ecklu-Mensah G, Choo-Kang C, Maseng MG, Donato S, Bovet P, Viswanathan B, Bedu-Addo K, Plange-Rhule J, Oti Boateng P, Forrester TE, Williams M, Lambert EV, Rae D, Sinyanya N, Luke A, Layden BT, O'Keefe S, Gilbert JA, Dugas LR. Gut microbiota and fecal short chain fatty acids differ with adiposity and country of origin: the METS-microbiome study. Nat Commun 2023; 14:5160. [PMID: 37620311 PMCID: PMC10449869 DOI: 10.1038/s41467-023-40874-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
The relationship between microbiota, short chain fatty acids (SCFAs), and obesity remains enigmatic. We employ amplicon sequencing and targeted metabolomics in a large (n = 1904) African origin cohort from Ghana, South Africa, Jamaica, Seychelles, and the US. Microbiota diversity and fecal SCFAs are greatest in Ghanaians, and lowest in Americans, representing each end of the urbanization spectrum. Obesity is significantly associated with a reduction in SCFA concentration, microbial diversity, and SCFA synthesizing bacteria, with country of origin being the strongest explanatory factor. Diabetes, glucose state, hypertension, obesity, and sex can be accurately predicted from the global microbiota, but when analyzed at the level of country, predictive accuracy is only universally maintained for sex. Diabetes, glucose, and hypertension are only predictive in certain low-income countries. Our findings suggest that adiposity-related microbiota differences differ between low-to-middle-income compared to high-income countries. Further investigation is needed to determine the factors driving this association.
Collapse
Affiliation(s)
- Gertrude Ecklu-Mensah
- Department of Pediatrics, Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Candice Choo-Kang
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
| | - Maria Gjerstad Maseng
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Dep. of Gastroenterology, Oslo University Hospital, Oslo, Norway
- Bio-Me, Oslo, Norway
| | - Sonya Donato
- Department of Pediatrics, Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Pascal Bovet
- University Center for Primary Care and Public Health (Unisanté), Lausanne University Hospital, Lausanne, Switzerland
- Ministry of Health, Victoria, Republic of Seychelles
| | | | - Kweku Bedu-Addo
- Department of Physiology, SMS, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jacob Plange-Rhule
- Department of Physiology, SMS, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Prince Oti Boateng
- Department of Physiology, SMS, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Terrence E Forrester
- Solutions for Developing Countries, University of the West Indies, Mona, Kingston, Jamaica
| | - Marie Williams
- Solutions for Developing Countries, University of the West Indies, Mona, Kingston, Jamaica
| | - Estelle V Lambert
- Research Unit for Exercise Science and Sports Medicine, University of Cape Town, Cape Town, South Africa
| | - Dale Rae
- Research Unit for Exercise Science and Sports Medicine, University of Cape Town, Cape Town, South Africa
| | - Nandipha Sinyanya
- Research Unit for Exercise Science and Sports Medicine, University of Cape Town, Cape Town, South Africa
| | - Amy Luke
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
| | - Brian T Layden
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Stephen O'Keefe
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jack A Gilbert
- Department of Pediatrics, Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Lara R Dugas
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA.
- Division of Epidemiology and Biostatistics, School of Public Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
5
|
Murga-Garrido SM, Ulloa-Pérez EJ, Díaz-Benítez CE, Orbe-Orihuela YC, Cornejo-Granados F, Ochoa-Leyva A, Sanchez-Flores A, Cruz M, Castañeda-Márquez AC, Plett-Torres T, Burguete García AI, Lagunas-Martínez A. Virulence Factors of the Gut Microbiome Are Associated with BMI and Metabolic Blood Parameters in Children with Obesity. Microbiol Spectr 2023; 11:e0338222. [PMID: 36786619 PMCID: PMC10101034 DOI: 10.1128/spectrum.03382-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/29/2023] [Indexed: 02/15/2023] Open
Abstract
The development of metabolic diseases is linked to the gut microbiota. A cross-sectional study involving 45 children (6 to 12 years old) was conducted to investigate the relationship between gut microbiota and childhood obesity. Anthropometric and metabolic measurements, food-frequency questionnaires (FFQs), and feces samples were obtained. Using the body mass index (BMI) z-score, we categorized each participant as normal weight (NW), or overweight and obese (OWOB). We determined 2 dietary profiles: one with complex carbohydrates and proteins (pattern 1), and the other with saturated fat and simple carbohydrates (pattern 2). The microbial taxonomic diversity and metabolic capacity were determined using shotgun metagenomics. We found differences between both BMI groups diversity. Taxa contributing to this difference, included Eubacterium sp., Faecalibacterium prausnitzii, Dialister, Monoglobus pectinilyticus, Bifidobacterium pseudocatenulatum, Intestinibacter bartlettii, Bacteroides intestinalis, Bacteroides uniformis, and Methanobrevibacter smithii. Metabolic capacity differences found between NW and OWOB, included the amino acid biosynthesis pathway, the cofactor, carrier, and vitamin biosynthesis pathway, the nucleoside and nucleotide biosynthesis and degradation pathways, the carbohydrate-sugar degradation pathway, and the amine and polyamine biosynthesis pathway. We found significant associations between taxa such as Ruminococcus, Mitsuokella multacida, Klebsiella variicola, and Citrobacter spp., metabolic pathways with the anthropometric, metabolic, and dietary data. We also found the microbiome's lipooligosaccharide (LOS) category as differentially abundant between BMI groups. Metabolic variations emerge during childhood as a result of complex nutritional and microbial interactions, which should be explained in order to prevent metabolic illnesses in adolescence and maturity. IMPORTANCE The alteration of gut microbiome composition has been commonly observed in diseases involving inflammation, such as obesity and metabolic impairment. Inflammatory host response in the gut can be a consequence of dietary driven dysbiosis. This response is conducive to blooms of particular bacterial species, adequate to survive in an inflammatory environment by means of genetical capability of utilizing alternative nutrients. Understanding the genomic and metabolic contribution of microbiota to inflammation, including virulence factor prevalence and functional potential, will contribute to identifying modifiable early life exposures and preventive strategies associated with obesity risk in childhood.
Collapse
Affiliation(s)
- S. M. Murga-Garrido
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
- PECEM (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - E. J. Ulloa-Pérez
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - C. E. Díaz-Benítez
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Y. C. Orbe-Orihuela
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - F. Cornejo-Granados
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - A. Ochoa-Leyva
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - A. Sanchez-Flores
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - M. Cruz
- Unidad de Investigación Médica en Bioquímica, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - A. C. Castañeda-Márquez
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - T. Plett-Torres
- PECEM (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - A. I. Burguete García
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - A. Lagunas-Martínez
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| |
Collapse
|
6
|
Shi F, Almerick T Boncan D, Wan HT, Chan TF, Zhang EL, Lai KP, Wong CKC. Hepatic metabolism gene expression and gut microbes in offspring, subjected to in-utero PFOS exposure and postnatal diet challenges. CHEMOSPHERE 2022; 308:136196. [PMID: 36041519 DOI: 10.1016/j.chemosphere.2022.136196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
We examined the changes in hepatic metabolic gene expression and gut microbiota of offspring exposed to PFOS in-utero. At GD17.5, our data showed that PFOS exposure decreased fetal bodyweights and hepatic metabolic gene expressions but increased relative liver mass and lipid accumulation. At PND21, in-utero high-dose PFOS-exposed offspring exhibited significantly greater bodyweight (catch-up-growth), associated with significant induction of hepatic metabolic gene expression. In addition, 16SrRNA-sequencing of the cecal samples revealed an increase in carbohydrate catabolism but a reduction in microbial polysaccharide synthesis and short-chain fatty acid (SCFA) metabolism. From PND21-80, a postnatal diet-challenge for the offspring was conducted. At PND80 under a normal diet, in-utero high-dose PFOS-exposed offspring maintained the growth "catch-up" effect. In contrast, in a high-fat-diet, the bodyweight of in-utero high-dose PFOS-exposed adult offspring were significantly lesser than the corresponding low-dose and control groups. Even though in the high-fat-diet, the in-utero PFOS-exposed adult offspring showed significant upregulation of hepatic metabolic genes, the lower bodyweight suggests that they had difficulty utilizing high-fat nutrients. Noteworthy, the metagenomic data showed a significant reduction in the biosynthesis of microbial polysaccharides, vitamin B, and SCFAs in the PFOS-exposed adult offspring. Furthermore, the observed effects were significantly reduced in the PFOS-exposed adult offspring with the high-fat diet but supplemented with sucrose. Our study demonstrated that in-utero PFOS exposure caused inefficient fat metabolism and increased the risk of hepatic steatosis in offspring.
Collapse
Affiliation(s)
- Feng Shi
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Delbert Almerick T Boncan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hin Ting Wan
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ting Fung Chan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eric L Zhang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong SAR, China
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Chris Kong-Chu Wong
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
7
|
D'Aloisio LD, Shetty V, Ballal M, Gibson DL. Following the Indian Immigrant: adoption of westernization results in a western gut microbiome and an increased risk of inflammatory bowel diseases. FEMS Microbiol Ecol 2022; 98:6825449. [PMID: 36370451 DOI: 10.1093/femsec/fiac133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
Indians who migrate to westernized countries such as Canada, the USA, and the UK are at an increased risk of developing inflammatory bowel disease (IBD). While the underlying aetiology of IBD remains unclear, a gut microbiome, i.e. no longer symbiotic with its host, is a major player. Increasing IBD incidence in Indian immigrants may be due to the adoption of western practices that result in loss of tolerance of a symbiotic community in the gut and its underlying immune responses. However, little is known about the microbial changes in the Indian gut, including shifts in the microbiome when they migrate to westernized countries. In this Current Opinion, we discuss what is known about the Indian gut microbiome and how living in a westernized environment may be impeding what was once a symbiotic relationship with their gut microbiome and intestinal mucosae, which may be the driving factor in their increased risk of IBD.
Collapse
Affiliation(s)
- Leah D D'Aloisio
- Department of Biology, University of British Columbia- Okanagan Campus, V1V 1V7 Kelowna, Canada
| | - Vignesh Shetty
- Enteric Disease Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, 576104 Manipal, India.,Department of Medicine, University of Cambridge, CB2 2QQ Cambridge, United Kingdom
| | - Mamatha Ballal
- Enteric Disease Division, Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, 576104 Manipal, India
| | - Deanna L Gibson
- Department of Biology, University of British Columbia- Okanagan Campus, V1V 1V7 Kelowna, Canada.,Department of Medicine, University of British Columbia- Okanagan Campus, V1V 1V7 Kelowna, Canada
| |
Collapse
|
8
|
Liang J, Li T, Zhao J, Wang C, Sun H. Current understanding of the human microbiome in glioma. Front Oncol 2022; 12:781741. [PMID: 36003766 PMCID: PMC9393498 DOI: 10.3389/fonc.2022.781741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
There is mounting evidence that the human microbiome is highly associated with a wide variety of central nervous system diseases. However, the link between the human microbiome and glioma is rarely noticed. The exact mechanism of microbiota to affect glioma remains unclear. Recent studies have demonstrated that the microbiome may affect the development, progress, and therapy of gliomas, including the direct impacts of the intratumoral microbiome and its metabolites, and the indirect effects of the gut microbiome and its metabolites. Glioma-related microbiome (gut microbiome and intratumoral microbiome) is associated with both tumor microenvironment and tumor immune microenvironment, which ultimately influence tumorigenesis, progression, and responses to treatment. In this review, we briefly summarize current knowledge regarding the role of the glioma-related microbiome, focusing on its gut microbiome fraction and a brief description of the intratumoral microbiome, and put forward the prospects in which microbiome can be applied in the future and some challenges still need to be solved.
Collapse
Affiliation(s)
- Jianhao Liang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Li
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiajia Zhao
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Laboratory Medicine, Clinical Biobank Center, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
- *Correspondence: Haitao Sun,
| |
Collapse
|
9
|
Liu L, Shah K. The Potential of the Gut Microbiome to Reshape the Cancer Therapy Paradigm: A Review. JAMA Oncol 2022; 8:1059-1067. [PMID: 35482355 DOI: 10.1001/jamaoncol.2022.0494] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance The gut microbiome, home to the vast kingdom of diverse commensal bacteria and other microorganisms residing within the gut, was once thought to only have roles primarily centered on digestive functions. However, recent advances in sequencing technology have elucidated intricate roles of the gut microbiome in cancer development and efficacy of therapeutic response that need to be comprehensively addressed from a clinically translational angle. Observations This review aims to highlight the current understanding of the association of the gut microbiome with the therapeutic response to immunotherapy, chemotherapy, radiotherapy, cancer surgery, and more, while also contextualizing possible synergistic strategies with the microbiome for tackling some of the most challenging tumors. It also provides insights on contemporary methods that target the microbiota and the current progression of findings being translated from bench to bedside. Conclusions and Relevance Ultimately, the importance of gut bacteria in cancer therapy cannot be overstated in its potential for ushering in a new era of cancer treatments. With the understanding that the microbiome may play critical roles in the tumor microenvironment, holistic approaches that integrate microbiome-modulating treatments with biological, immune, cell-based, and surgical cancer therapies should be explored.
Collapse
Affiliation(s)
- Longsha Liu
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, Massachusetts.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, Massachusetts.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
10
|
Abstract
The microbiota-gut-brain-axis (MGBA) is a bidirectional communication network between gut microbes and their host. Many environmental and host-related factors affect the gut microbiota. Dysbiosis is defined as compositional and functional alterations of the gut microbiota that contribute to the pathogenesis, progression and treatment responses to disease. Dysbiosis occurs when perturbations of microbiota composition and function exceed the ability of microbiota and its host to restore a symbiotic state. Dysbiosis leads to dysfunctional signaling of the MGBA, which regulates the development and the function of the host's immune, metabolic, and nervous systems. Dysbiosis-induced dysfunction of the MGBA is seen with aging and stroke, and is linked to the development of common stroke risk factors such as obesity, diabetes, and atherosclerosis. Changes in the gut microbiota are also seen in response to stroke, and may impair recovery after injury. This review will begin with an overview of the tools used to study the MGBA with a discussion on limitations and potential experimental confounders. Relevant MGBA components are introduced and summarized for a better understanding of age-related changes in MGBA signaling and its dysfunction after stroke. We will then focus on the relationship between the MGBA and aging, highlighting that all components of the MGBA undergo age-related alterations that can be influenced by or even driven by the gut microbiota. In the final section, the current clinical and preclinical evidence for the role of MGBA signaling in the development of stroke risk factors such as obesity, diabetes, hypertension, and frailty are summarized, as well as microbiota changes with stroke in experimental and clinical populations. We conclude by describing the current understanding of microbiota-based therapies for stroke including the use of pre-/pro-biotics and supplementations with bacterial metabolites. Ongoing progress in this new frontier of biomedical sciences will lead to an improved understanding of the MGBA's impact on human health and disease.
Collapse
Affiliation(s)
- Pedram Honarpisheh
- Department of Neurology, University of Texas McGovern Medical School, Houston (P.H., L.D.M.)
| | - Robert M Bryan
- Department of Anesthesiology, Baylor College of Medicine, Houston, TX (R.M.B.)
| | - Louise D McCullough
- Department of Neurology, University of Texas McGovern Medical School, Houston (P.H., L.D.M.)
| |
Collapse
|
11
|
Couchet M, Breuillard C, Corne C, Rendu J, Morio B, Schlattner U, Moinard C. Ornithine Transcarbamylase - From Structure to Metabolism: An Update. Front Physiol 2021; 12:748249. [PMID: 34658931 PMCID: PMC8517447 DOI: 10.3389/fphys.2021.748249] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
Ornithine transcarbamylase (OTC; EC 2.1.3.3) is a ubiquitous enzyme found in almost all organisms, including vertebrates, microorganisms, and plants. Anabolic, mostly trimeric OTCs catalyze the production of L-citrulline from L-ornithine which is a part of the urea cycle. In eukaryotes, such OTC localizes to the mitochondrial matrix, partially bound to the mitochondrial inner membrane and part of channeling multi-enzyme assemblies. In mammals, mainly two organs express OTC: the liver, where it is an integral part of the urea cycle, and the intestine, where it synthesizes citrulline for export and plays a major role in amino acid homeostasis, particularly of L-glutamine and L-arginine. Here, we give an overview on OTC genes and proteins, their tissue distribution, regulation, and physiological function, emphasizing the importance of OTC and urea cycle enzymes for metabolic regulation in human health and disease. Finally, we summarize the current knowledge of OTC deficiency, a rare X-linked human genetic disorder, and its emerging role in various chronic pathologies.
Collapse
Affiliation(s)
- Morgane Couchet
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | - Charlotte Breuillard
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| | | | - John Rendu
- Centre Hospitalier Université Grenoble Alpes, Grenoble, France
| | - Béatrice Morio
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Lyon, France
| | - Uwe Schlattner
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France.,Institut Universitaire de France, Paris, France
| | - Christophe Moinard
- Université Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics, Grenoble, France
| |
Collapse
|
12
|
D’Alessandro A, Thomas T, Akpan IJ, Reisz JA, Cendali FI, Gamboni F, Nemkov T, Thangaraju K, Katneni U, Tanaka K, Kahn S, Wei AZ, Valk JE, Hudson KE, Roh D, Moriconi C, Zimring JC, Hod EA, Spitalnik SL, Buehler PW, Francis RO. Biological and Clinical Factors Contributing to the Metabolic Heterogeneity of Hospitalized Patients with and without COVID-19. Cells 2021; 10:2293. [PMID: 34571942 PMCID: PMC8467961 DOI: 10.3390/cells10092293] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
The Corona Virus Disease 2019 (COVID-19) pandemic represents an ongoing worldwide challenge. The present large study sought to understand independent and overlapping metabolic features of samples from acutely ill patients (n = 831) that tested positive (n = 543) or negative (n = 288) for COVID-19. High-throughput metabolomics analyses were complemented with antigen and enzymatic activity assays on plasma from acutely ill patients collected while in the emergency department, at admission, or during hospitalization. Lipidomics analyses were also performed on COVID-19-positive or -negative subjects with the lowest and highest body mass index (n = 60/group). Significant changes in amino acid and fatty acid/acylcarnitine metabolism emerged as highly relevant markers of disease severity, progression, and prognosis as a function of biological and clinical variables in these patients. Further, machine learning models were trained by entering all metabolomics and clinical data from half of the COVID-19 patient cohort and then tested on the other half, yielding ~78% prediction accuracy. Finally, the extensive amount of information accumulated in this large, prospective, observational study provides a foundation for mechanistic follow-up studies and data sharing opportunities, which will advance our understanding of the characteristics of the plasma metabolism in COVID-19 and other acute critical illnesses.
Collapse
Affiliation(s)
- Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA; (J.A.R.); (F.I.C.); (F.G.); (T.N.)
| | - Tiffany Thomas
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - Imo J. Akpan
- Division of Hematology/Oncology, Department of Medicine, Irving Medical Center, Columbia University, New York, NY 10032, USA; (I.J.A.); (S.K.); (A.Z.W.)
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA; (J.A.R.); (F.I.C.); (F.G.); (T.N.)
| | - Francesca I. Cendali
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA; (J.A.R.); (F.I.C.); (F.G.); (T.N.)
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA; (J.A.R.); (F.I.C.); (F.G.); (T.N.)
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA; (J.A.R.); (F.I.C.); (F.G.); (T.N.)
| | - Kiruphagaran Thangaraju
- Center for Blood Oxygen Transport, Department of Pathology, Department of Pediatrics, University of Maryland, Baltimore, MD 21201, USA; (K.T.); (U.K.); (P.W.B.)
| | - Upendra Katneni
- Center for Blood Oxygen Transport, Department of Pathology, Department of Pediatrics, University of Maryland, Baltimore, MD 21201, USA; (K.T.); (U.K.); (P.W.B.)
| | - Kenichi Tanaka
- Department of Anesthesiology, University of Maryland, Baltimore, MD 21201, USA;
- Department of Anesthesiology, University of Oklahoma College of Medicine, Oklahoma City, OK 73126-0901, USA
| | - Stacie Kahn
- Division of Hematology/Oncology, Department of Medicine, Irving Medical Center, Columbia University, New York, NY 10032, USA; (I.J.A.); (S.K.); (A.Z.W.)
| | - Alexander Z. Wei
- Division of Hematology/Oncology, Department of Medicine, Irving Medical Center, Columbia University, New York, NY 10032, USA; (I.J.A.); (S.K.); (A.Z.W.)
| | - Jacob E. Valk
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - Krystalyn E. Hudson
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - David Roh
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA;
| | - Chiara Moriconi
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - James C. Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA 22903, USA;
| | - Eldad A. Hod
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - Steven L. Spitalnik
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| | - Paul W. Buehler
- Center for Blood Oxygen Transport, Department of Pathology, Department of Pediatrics, University of Maryland, Baltimore, MD 21201, USA; (K.T.); (U.K.); (P.W.B.)
| | - Richard O. Francis
- Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; (T.T.); (J.E.V.); (K.E.H.); (C.M.); (E.A.H.); (S.L.S.); (R.O.F.)
| |
Collapse
|
13
|
Citrulline, Biomarker of Enterocyte Functional Mass and Dietary Supplement. Metabolism, Transport, and Current Evidence for Clinical Use. Nutrients 2021; 13:nu13082794. [PMID: 34444954 PMCID: PMC8398474 DOI: 10.3390/nu13082794] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
L-Citrulline is a non-essential but still important amino acid that is released from enterocytes. Because plasma levels are reduced in case of impaired intestinal function, it has become a biomarker to monitor intestinal integrity. Moreover, oxidative stress induces protein citrullination, and antibodies against anti-citrullinated proteins are useful to monitor rheumatoid diseases. Citrullinated histones, however, may even predict a worse outcome in cancer patients. Supplementation of citrulline is better tolerated compared to arginine and might be useful to slightly improve muscle strength or protein balance. The following article shall provide an overview of L-citrulline properties and functions, as well as the current evidence for its use as a biomarker or as a therapeutic supplement.
Collapse
|
14
|
Johnson SD, Olwenyi OA, Bhyravbhatla N, Thurman M, Pandey K, Klug EA, Johnston M, Dyavar SR, Acharya A, Podany AT, Fletcher CV, Mohan M, Singh K, Byrareddy SN. Therapeutic implications of SARS-CoV-2 dysregulation of the gut-brain-lung axis. World J Gastroenterol 2021; 27:4763-4783. [PMID: 34447225 PMCID: PMC8371510 DOI: 10.3748/wjg.v27.i29.4763] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
The emergence and rapid spread of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused over 180 million confirmed cases resulting in over 4 million deaths worldwide with no clear end in sight for the coronavirus disease 19 (COVID-19) pandemic. Most SARS-CoV-2 exposed individuals experience mild to moderate symptoms, including fever, cough, fatigue, and loss of smell and taste. However, many individuals develop pneumonia, acute respiratory distress syndrome, septic shock, and multiorgan dysfunction. In addition to these primarily respiratory symptoms, SARS-CoV-2 can also infiltrate the central nervous system, which may damage the blood-brain barrier and the neuron's synapses. Resultant inflammation and neurodegeneration in the brain stem can further prevent efferent signaling to cranial nerves, leading to the loss of anti-inflammatory signaling and normal respiratory and gastrointestinal functions. Additionally, SARS-CoV-2 can infect enterocytes resulting in gut damage followed by microbial dysbiosis and translocation of bacteria and their byproducts across the damaged epithelial barrier. As a result, this exacerbates pro-inflammatory responses both locally and systemically, resulting in impaired clinical outcomes. Recent evidence has highlighted the complex interactions that mutually modulate respiratory, neurological, and gastrointestinal function. In this review, we discuss the ways SARS-CoV-2 potentially disrupts the gut-brain-lung axis. We further highlight targeting specific responses to SARS-CoV-2 for the development of novel, urgently needed therapeutic interventions. Finally, we propose a prospective related to the individuals from Low- and Middle-Income countries. Here, the underlying propensity for heightened gut damage/microbial translocation is likely to result in worse clinical outcomes during this COVID-19 pandemic.
Collapse
Affiliation(s)
- Samuel D Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Omalla A Olwenyi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Namita Bhyravbhatla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Michellie Thurman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Kabita Pandey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Elizabeth A Klug
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Morgan Johnston
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Shetty Ravi Dyavar
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, University of Nebraska Medical Center (UNMC) Center for Drug Discovery, Omaha, NE 68198, United States
| | - Mahesh Mohan
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, United States
| | - Kamal Singh
- Department of Molecular Microbiology and Immunology and Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| |
Collapse
|
15
|
Giron LB, Dweep H, Yin X, Wang H, Damra M, Goldman AR, Gorman N, Palmer CS, Tang HY, Shaikh MW, Forsyth CB, Balk RA, Zilberstein NF, Liu Q, Kossenkov A, Keshavarzian A, Landay A, Abdel-Mohsen M. Plasma Markers of Disrupted Gut Permeability in Severe COVID-19 Patients. Front Immunol 2021; 12:686240. [PMID: 34177935 PMCID: PMC8219958 DOI: 10.3389/fimmu.2021.686240] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
A disruption of the crosstalk between the gut and the lung has been implicated as a driver of severity during respiratory-related diseases. Lung injury causes systemic inflammation, which disrupts gut barrier integrity, increasing the permeability to gut microbes and their products. This exacerbates inflammation, resulting in positive feedback. We aimed to test whether severe Coronavirus disease 2019 (COVID-19) is associated with markers of disrupted gut permeability. We applied a multi-omic systems biology approach to analyze plasma samples from COVID-19 patients with varying disease severity and SARS-CoV-2 negative controls. We investigated the potential links between plasma markers of gut barrier integrity, microbial translocation, systemic inflammation, metabolome, lipidome, and glycome, and COVID-19 severity. We found that severe COVID-19 is associated with high levels of markers of tight junction permeability and translocation of bacterial and fungal products into the blood. These markers of disrupted intestinal barrier integrity and microbial translocation correlate strongly with higher levels of markers of systemic inflammation and immune activation, lower levels of markers of intestinal function, disrupted plasma metabolome and glycome, and higher mortality rate. Our study highlights an underappreciated factor with significant clinical implications, disruption in gut functions, as a potential force that may contribute to COVID-19 severity.
Collapse
Affiliation(s)
- Leila B Giron
- The Wistar Institute, Philadelphia, PA, United States
| | - Harsh Dweep
- The Wistar Institute, Philadelphia, PA, United States
| | - Xiangfan Yin
- The Wistar Institute, Philadelphia, PA, United States
| | - Han Wang
- The Wistar Institute, Philadelphia, PA, United States
| | | | | | - Nicole Gorman
- The Wistar Institute, Philadelphia, PA, United States
| | - Clovis S Palmer
- The Burnet Institute, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - Hsin-Yao Tang
- The Wistar Institute, Philadelphia, PA, United States
| | - Maliha W Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University, Chicago, IL, United States
| | - Christopher B Forsyth
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University, Chicago, IL, United States.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Robert A Balk
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Netanel F Zilberstein
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Qin Liu
- The Wistar Institute, Philadelphia, PA, United States
| | | | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University, Chicago, IL, United States.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Alan Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | | |
Collapse
|
16
|
Evaluation of the Mechanisms Underlying Amino Acid and Microbiota Interactions in Intestinal Infections Using Germ-Free Animals. INFECTIOUS MICROBES AND DISEASES 2021. [DOI: 10.1097/im9.0000000000000060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Kashyap S, Shivakumar N, Sejian V, Deutz NEP, Preston T, Sreeman S, Devi S, Kurpad AV. Goat milk protein digestibility in relation to intestinal function. Am J Clin Nutr 2021; 113:845-853. [PMID: 33677496 PMCID: PMC8023838 DOI: 10.1093/ajcn/nqaa400] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Milk is an important high-quality animal protein source in low- and middle-income countries (LMICs). Although the true ileal digestibility and absorption of milk has been shown to be high in French adults, this may be lower in individuals from LMICs who are at risk of environmental enteropathy. OBJECTIVE To determine the true ileal indispensable amino acid (IAA) digestibility of intrinsically labeled goat milk protein in South Indian women of reproductive age (WRA), using the dual-isotope tracer technique, and to measure intestinal absorption of amino acid and inert sugar in the same participants using L-allo-isoleucine and a dual-sugar assay. METHODS Milk with 2H-labeled protein collected from a lactating goat fed intrinsically 2H-labeled fodder (maize and cowpea) was spray dried. Labeled milk protein was administered in a plateau feeding protocol to WRA with normal BMI, in whom urinary lactulose and mannitol recovery and the lactulose/mannitol ratio (LMR) were measured, to determine its true ileal IAA digestibility by the dual-isotope tracer technique with a reference U-13C-amino acid mixture. A phenylalanine absorption index was calculated from the plasma to meal ratio of 13C9 phenylalanine within the digestibility protocol. On a separate day, the allo-isoleucine absorption index was estimated from the ratio of plasma allo-isoleucine enrichments after oral 13C6-15N-L- and intravenous 2H10-L-allo-isoleucine administration. RESULTS The means ± SDs of true ileal IAA digestibility of goat milk protein, lactulose and mannitol recovery, LMR, allo-isoleucine and phenylalanine absorption index were 94.0 ± 2.9%, 0.09 ± 0.03%, 7.9 ± 2.3%, 0.012 ± 0.004, 88.4 ± 3.8% and 24.5 ± 1.6%, respectively. The LMR correlated with the allo-isoleucine absorption index (rs = -0.93, P = 0.008). CONCLUSION The true ileal digestibility of goat milk protein in South Indian WRA with normal intestinal absorptive function and integrity was comparable to earlier estimates in healthy French adults.
Collapse
Affiliation(s)
- Sindhu Kashyap
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Nirupama Shivakumar
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Veerasamy Sejian
- ICAR—National Institute of Animal Nutrition and Physiology, Adugodi, Bangalore, India
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging and Longevity, Department of Health and Kinesiology, Texas A&M University, College Station, TX, USA
| | - Thomas Preston
- Scottish Universities Environmental Research Centre, East Kilbride, Scotland, UK
| | - Sheshshayee Sreeman
- Department of Crop Physiology, University of Agricultural Sciences, Bangalore, India
| | - Sarita Devi
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
| | - Anura V Kurpad
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, India
- Department of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| |
Collapse
|
18
|
Si H, Han Y, Liu H, Lou Y, Li Z. Effects of rumen-protected arginine supplementation on the plasma amino acids and gut microbiota of sika deer (Cervus nippon). Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.114828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
19
|
Diet- and sex-related changes of gut microbiota composition and functional profiles after 4 months of weight loss intervention. Eur J Nutr 2021; 60:3279-3301. [PMID: 33591390 DOI: 10.1007/s00394-021-02508-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Obesity has been related to intestinal dysbiosis and the modification of gut microbiota composition by dietary strategies becomes a promising strategy to help manage obesity. The aim of the current study was to evaluate the effect of two weight-loss diets on the composition and functional profile of gut microbiota. METHODS 55 men and 124 women with BMI > 25 kg/m2 were randomly assigned to moderately high-protein (MHP) or low-fat (LF) diet. Differences in fecal bacteria abundance (based on 16 s rRNA sequencing) between before and after 4 months of calorie restriction was analyzed using EdgeR tool in MicrobiomeAnalyst platform. Bacterial functional profile was predicted using Tax4Fun and metagenomeSeq analysis. Significant KEGG Orthology (KO) terms were selected for the metabolomic study using chromatography. RESULTS After the intervention, MHP-men showed a significant decrease in Negativicutes, Selenomonadales, Dielma and Dielma fastidiosa. LF-men showed a significant increase in Bacilli, Lactobacillales, Christensenellaceae, Peptococcaceae, and Streptococcaceae, Peptococcus, Streptococcus and Christensenella, Duncaniella dubosii_CP039396_93.49%, Roseburia sp_AB744234_98.96% and Alistipes inops_KJ572413_99.57%. MHP-women increased Pasteurellales, Phascolarctobacterium succinatutens, Ruthenibacterium lactatiformans_LR215981_99.55% and decreased in Phascolarctobacterium succinatutens_NR112902_99.56%. Finally, LF-women presented a significant decrease in Bacteroides clarus and Erysipelothrix inopinata_CP060715_84.4%. Surprisingly, no matching bacterial changes were found between these four groups. A total of 42 KO, 10 metabolic pathways and 107 related metabolites related were found implicated in these bacterial changes. Seven metabolites were confirmed in plasma. CONCLUSION Weight-loss-related-changes in gut microbiome composition and the functional profile occur in a sex- and diet-related manner, showing that women and men could differentially benefit from the consumption of MHP and LF diets. TRIAL REGISTRATION NCT02737267, 10th March 2016 retrospectively registered.
Collapse
|
20
|
Si H, Liu H, Nan W, Li G, Li Z, Lou Y. Effects of Arginine Supplementation on Serum Metabolites and the Rumen Bacterial Community of Sika Deer ( Cervus nippon). Front Vet Sci 2021; 8:630686. [PMID: 33614769 PMCID: PMC7892468 DOI: 10.3389/fvets.2021.630686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/13/2021] [Indexed: 11/20/2022] Open
Abstract
Velvet antler is a regeneration organ of sika deer (Cervus nippon) and an important Chinese medicine, and nutrient metabolism affects its growth. Here, we investigated the effects of arginine supplementation on antler growth, serum biochemical indices, and the rumen bacterial community of sika deer during the antler growth period. Fifteen male sika deer (6 years old) were randomly assigned to three dietary groups, which were supplemented with 0 (n = 5, CON), 2.5 (n = 5, LArg), or 5.0 g/d (n = 5, HArg) L-arginine. The IGF-1, ALT and AST concentrations in the serum of LArg sika deer were significantly higher than those in the serum of CON (P < 0.05) and HArg deer (P < 0.05). The phyla Bacteroidetes, Firmicutes, and Proteobacteria were dominant in the rumen of sika deer among the three groups. Comparison of alpha diversities showed that the ACE and Chao1 indices significantly increased in the LArg and HArg groups compared with those in the CON group. PCoA and ANOSIM results showed that the bacterial community was significantly changed between the CON and LArg groups. Moreover, the relative abundances of Fibrobacter spp. and Prevotellaceae UCG-003 increased, but those of Clostridium sensu stricto 1 and Corynebacterium 1 decreased in the LArg and HArg groups compared with those in the CON group. Additionally, the relative abundances of 19 OTUs were significantly different between the LArg and HArg groups. These results revealed that arginine supplementation affected the sika deer rumen bacterial community and serum biochemical indices.
Collapse
Affiliation(s)
- Huazhe Si
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.,Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Hanlu Liu
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weixiao Nan
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guangyu Li
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhipeng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.,Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yujie Lou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
21
|
Bandyopadhyay S, Shivakumar N, Kurpad AV. Protein intakes of pregnant women and children in India-protein quality implications. MATERNAL & CHILD NUTRITION 2020; 16 Suppl 3:e12952. [PMID: 33347716 PMCID: PMC7752127 DOI: 10.1111/mcn.12952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/25/2019] [Accepted: 12/29/2019] [Indexed: 11/28/2022]
Abstract
The recent National Family Health Survey (NFHS-4, 2016) reports a national average of 18% for low birthweight (LBW) and 38% for stunting in children <5 years. Nutrition and environmental influences (chronic enteric pathogenic exposure through poor water, sanitation, and hygiene) are two critical factors that impact the health outcomes of the populxation. This is particularly relevant for vulnerable age groups such as pregnant women and children <5 years, who bear long-lasting and intergenerational consequences of impoverished nutrition and suboptimal living conditions. The present review provides, for the first time, an analysis of indispensable amino acid (IAA) requirements for pregnant women, separately for the second and third trimesters, using protein accretion data from a recent Indian study. Furthermore, using these estimates for pregnancy, and the current IAA requirements for young children, the quality of protein was assessed in Indian diets consumed by pregnant women and children (1-3 and 4-6 years) from national representative rural National Nutrition Monitoring Bureau survey. The assessment was considered in the context of an adverse environment and in relation to outcomes such as LBW, stunting, and underweight. Finally, an assessment was made of the proportion of the surveyed population at risk of dietary quality protein inadequacy and implications for planning nutrition intervention programmes. Specifically, state-wise estimates of the risk of quality protein inadequacy are provided, in addition to evaluations of additional dietary supplementation, which could inform the policy of supplementary nutrition programmes to improve health outcomes.
Collapse
Affiliation(s)
- Sulagna Bandyopadhyay
- Division of Nutrition, St. John's Research InstituteSt. John's National Academy of Health SciencesSarjapur Road, Bangalore560034India
| | - Nirupama Shivakumar
- Division of Nutrition, St. John's Research InstituteSt. John's National Academy of Health SciencesSarjapur Road, Bangalore560034India
| | - Anura V. Kurpad
- Division of Nutrition, St. John's Research InstituteSt. John's National Academy of Health SciencesSarjapur Road, Bangalore560034India
- Department of Physiology, St. John's Medical CollegeSt. John's National Academy of Health SciencesSarjapur Road, Bangalore560034India
| |
Collapse
|
22
|
Dehhaghi M, Kazemi Shariat Panahi H, Heng B, Guillemin GJ. The Gut Microbiota, Kynurenine Pathway, and Immune System Interaction in the Development of Brain Cancer. Front Cell Dev Biol 2020; 8:562812. [PMID: 33330446 PMCID: PMC7710763 DOI: 10.3389/fcell.2020.562812] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
Human gut microbiota contains a large, complex, dynamic microbial community of approximately 1014 microbes from more than 1,000 microbial species, i.e., equivalent to 4 × 106 genes. Numerous evidence links gut microbiota with human health and diseases. Importantly, gut microbiota is involved in the development and function of the brain through a bidirectional pathway termed as the gut-brain axis. Interaction between gut microbiota and immune responses can modulate the development of neuroinflammation and cancer diseases in the brain. With respect of brain cancer, gut microbiota could modify the levels of antioxidants, amyloid protein and lipopolysaccharides, arginase 1, arginine, cytochrome C, granulocyte-macrophage colony-stimulating factor signaling (GM-CSF), IL-4, IL-6, IL-13, IL-17A, interferon gamma (IFN-γ), reactive oxygen species (ROS), reactive nitrogen species (e.g., nitric oxide and peroxynitrite), short-chain fatty acids (SCFAs), tryptophan, and tumor necrosis factor-β (TGF-β). Through these modifications, gut microbiota can modulate apoptosis, the aryl hydrocarbon receptor (AhR), autophagy, caspases activation, DNA integrity, microglia dysbiosis, mitochondria permeability, T-cell proliferation and functions, the signal transducer and activator of transcription (STAT) pathways, and tumor cell proliferation and metastasis. The outcome of such interventions could be either oncolytic or oncogenic. This review scrutinizes the oncogenic and oncolytic effects of gut microbiota by classifying the modification mechanisms into (i) amino acid deprivation (arginine and tryptophan); (ii) kynurenine pathway; (iii) microglia dysbiosis; and (iv) myeloid-derived suppressor cells (MDSCs). By delineating the complexity of the gut-microbiota-brain-cancer axis, this review aims to help the research on the development of novel therapeutic strategies that may aid the efficient eradication of brain cancers.
Collapse
Affiliation(s)
- Mona Dehhaghi
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Pandis Community, Sydney, NSW, Australia.,Department of Microbial Biotechnology, School of Biology and Centre of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Hamed Kazemi Shariat Panahi
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Microbial Biotechnology, School of Biology and Centre of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Benjamin Heng
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Pandis Community, Sydney, NSW, Australia
| |
Collapse
|
23
|
Gut Prevotella as a possible biomarker of diet and its eubiotic versus dysbiotic roles: a comprehensive literature review. Br J Nutr 2019; 122:131-140. [PMID: 30924428 DOI: 10.1017/s0007114519000680] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The gut microbiota has a profound impact on human health. Emerging data show that dietary patterns are associated with different communities of bacterial species within the gut. Prevotella species have been correlated with plant-rich diets, abundant in carbohydrates and fibres. Dysbiosis within the gut ecosystem has been associated with the development of non-communicable diseases such as obesity, the metabolic syndrome, inflammatory bowel disease, irritable bowel syndrome, colorectal cancer, type 1 diabetes, allergies and other diseases. The purpose of this comprehensive literature review was to evaluate the available data on the impact of diet on the Prevotella genus, as a dietary fibre fermenter in the gut as well as its implications as a potential biomarker for homeostasis or disease state through its metabolite signature. Studies were identified by conducting PubMed, Web of Science Core Collection and Google Scholar electronic searches. We found eighty-five publications reporting the impact of dietary patterns on gut microbial communities, including Prevotella or Prevotella/Bacteroides ratio in particular. Moreover, the role of Prevotella species on health status was also evaluated. Prevotella possess a high genetic diversity, representing one of the important groups found in the oral cavity and large intestine of man. The gut commensal Prevotella bacteria contribute to polysaccharide breakdown, being dominant colonisers of agrarian societies. However, studies also suggested a potential role of Prevotella species as intestinal pathobionts. Further metagenomic studies are needed in order to reveal health- or disease-modulating properties of Prevotella species in the gut.
Collapse
|
24
|
Kuriyan R, Naqvi S, Bhat KG, Thomas T, Thomas A, George S, Nagarajarao SC, Sachdev HS, Preston T, Kurpad AV. Estimation of protein requirements in Indian pregnant women using a whole-body potassium counter. Am J Clin Nutr 2019; 109:1064-1070. [PMID: 30982859 PMCID: PMC6517275 DOI: 10.1093/ajcn/nqz011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/18/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The 2007 World Health Organization/Food and Agriculture Organization/United Nations University (WHO/FAO/UNU) recommendation for the Estimated Average Requirement (EAR) of additional protein during pregnancy for a gestational weight gain (GWG) of 12 kg (recalculated from a GWG of 13.8 kg) is 6.7 and 21.7 g/d in the second and the third trimester, respectively. This EAR is based on measurements of potassium accretion in high-income country (HIC) pregnant women. It is not known if low- to middle-income country, but well-nourished, pregnant women have comparable requirements. OBJECTIVE We aimed to estimate total body potassium (TBK) accretion during pregnancy in Indian pregnant women, using a whole-body potassium counter (WBKC), to measure their additional protein EAR. METHODS Well-nourished pregnant women (20-40 y, n = 38, middle socioeconomic stratum) were recruited in the first trimester of pregnancy. Anthropometric, dietary, and physical activity measurements, and measurements of TBK using a WBKC, were performed at each trimester and at birth. RESULTS The mid-trimester weight gain was 2.7 kg and 8.0 kg in the second and the third trimester, respectively, for an average 37-wk GWG of 10.7 kg and a mean birth weight of 3.0 kg. Protein accretion was 2.7 and 5.7 g/d, for an EAR of 8.2 and 18.9 g/d in the second and the third trimester, respectively. The additional protein EAR, calculated for a GWG of 12 kg, was 9.1 and 21.2 g/d in the second and the third trimester, respectively. CONCLUSION The additional protein requirements of well-nourished Indian pregnant women for a GWG of 12 kg in the second and third trimesters were similar to the recalculated 2007 WHO/FAO/UNU requirements for 12 kg.
Collapse
Affiliation(s)
- Rebecca Kuriyan
- Division of Nutrition, St. John’s Research Institute, Bengaluru, India
| | - Saba Naqvi
- Division of Nutrition, St. John’s Research Institute, Bengaluru, India
| | - Kishor G Bhat
- Division of Nutrition, St. John’s Research Institute, Bengaluru, India
| | - Tinku Thomas
- Division of Epidemiology and Biostatistics, St. John’s Research Institute, Bengaluru, India
| | - Annamma Thomas
- Department of Obstetrics and Gynecology, St. John’s Medical College Hospital, Bengaluru, India
| | - Shirley George
- Department of Obstetrics and Gynecology, St. John’s Medical College Hospital, Bengaluru, India
| | - Sheela C Nagarajarao
- Department of Obstetrics and Gynecology, St. John’s Medical College Hospital, Bengaluru, India
| | | | - Thomas Preston
- Stable Isotope Biochemistry Laboratory, Scottish Universities Environmental Research Centre, Glasgow, United Kingdom
| | - Anura V Kurpad
- Department of Physiology, St. John’s Medical College, Bengaluru, India
| |
Collapse
|
25
|
Kashyap S, Shivakumar N, Varkey A, Duraisamy R, Thomas T, Preston T, Devi S, Kurpad AV. Ileal digestibility of intrinsically labeled hen's egg and meat protein determined with the dual stable isotope tracer method in Indian adults. Am J Clin Nutr 2018; 108:980-987. [PMID: 30272112 PMCID: PMC6250983 DOI: 10.1093/ajcn/nqy178] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/04/2018] [Indexed: 01/18/2023] Open
Abstract
Background Protein quality assessment through the Digestible Indispensable Amino Acid Score requires accurate measurements of true ileal protein and amino acid digestibility, for which a dual isotope technique was recently developed. However, the ileal digestibility of indispensable amino acids (IAA) in humans from high-quality proteins is not well known. Objective The aim of this study was to intrinsically label hen's egg and meat protein by the use of uniformly 2H-labeled amino acids, and to measure their true ileal indispensable amino acid (IAA) digestibility via the dual isotope method in humans. Design 2H-labeled lyophilized boiled egg white protein, whole boiled egg, and cooked meat were obtained from layer hens (BV-300) administered a uniformly 2H-labeled amino acid mix orally for 35 d with their daily feed. The ileal IAA digestibility of these proteins was determined with reference to digestibility of previously characterized [U-13C]spirulina in a dual tracer method in healthy Indian subjects whose intestinal health was measured by the plasma kynurenine-to-tryptophan (KT) ratio. Results All subjects had normal KT ratios. The mean ± SD true ileal IAA digestibility of 2H-labeled egg white protein, whole boiled egg, and cooked meat was 86.3% ± 4.6%, 89.4% ± 4.5%, and 92.0% ± 2.8%, respectively. Leucine digestibility correlated with the KT ratio (r = -0.772; P = 0.009). Conclusions Uniformly 2H-labeled hen's egg and meat protein can be used to measure ileal IAA digestibility by the dual isotope tracer approach in humans. The mean IAA digestibility values for these high-quality proteins in the healthy Indians studied were similar to values obtained in earlier human and animal experiments. Leucine digestibility in these meal matrices correlated with the KT ratio, but this aspect needs further evaluation. This trial was registered at the Clinical Trials Registry of India (http://ctri.nic.in) as CTRI/2018/03/012265.
Collapse
Affiliation(s)
- Sindhu Kashyap
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Nirupama Shivakumar
- Departments of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Aneesia Varkey
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Rajendran Duraisamy
- Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Adugodi, Bangalore, India
| | - Tinku Thomas
- Departments of Biostatistics, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Thomas Preston
- Scottish Universities Environmental Research Centre, East Kilbride, Scotland, United Kingdom
| | - Sarita Devi
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Anura V Kurpad
- Division of Nutrition, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India,Departments of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India,Address correspondence to AVK (e-mail: )
| |
Collapse
|
26
|
Nguyen TL, Chun WK, Kim A, Kim N, Roh HJ, Lee Y, Yi M, Kim S, Park CI, Kim DH. Dietary Probiotic Effect of Lactococcus lactis WFLU12 on Low-Molecular-Weight Metabolites and Growth of Olive Flounder ( Paralichythys olivaceus). Front Microbiol 2018; 9:2059. [PMID: 30233536 PMCID: PMC6134039 DOI: 10.3389/fmicb.2018.02059] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 08/13/2018] [Indexed: 01/21/2023] Open
Abstract
The use of probiotics is considered an attractive biocontrol method. It is effective in growth promotion in aquaculture. However, the mode of action of probiotics in fish in terms of growth promotion remains unclear. The objective of the present study was to investigate growth promotion effect of dietary administration of host-derived probiotics, Lactococcus lactis WFLU12, on olive flounder compared to control group fed with basal diet by analyzing their intestinal and serum metabolome using capillary electrophoresis mass spectrometry with time-of flight (CE-TOFMS). Results of CE-TOFMS revealed that 53 out of 200 metabolites from intestinal luminal metabolome and 5 out of 171 metabolites from serum metabolome, respectively, were present in significantly higher concentrations in the probiotic-fed group than those in the control group. Concentrations of metabolites such as citrulline, tricarboxylic acid cycle (TCA) intermediates, short chain fatty acids, vitamins, and taurine were significantly higher in the probiotic-fed group than those in the control group. The probiotic strain WFLU12 also possesses genes encoding enzymes to help produce these metabolites. Therefore, it is highly likely that these increased metabolites linked to growth promotion in olive flounder are due to supplementation of the probiotic strain. To the best of our knowledge, this is the first study to show that dietary probiotics can greatly influence metabolome in fish. Findings of the present study may reveal important implications for maximizing the efficiency of using dietary additives to optimize fish health and growth.
Collapse
Affiliation(s)
- Thanh Luan Nguyen
- Department of Veterinary Medicine, HUTECH Institute of Applied Science, Ho Chi Minh City University of Technology, Ho Chi Minh City, Vietnam.,Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Won-Kyong Chun
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Ahran Kim
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Nameun Kim
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Heyong Jin Roh
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Yoonhang Lee
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| | - Myunggi Yi
- Department of Biomedical Engineering, College of Engineering, Pukyong National University, Busan, South Korea
| | - Suhkmann Kim
- Department of Chemistry, Center for Proteome Biophysics, Chemistry Institute for Functional Materials, Pusan National University, Busan, South Korea
| | - Chan-Il Park
- Department of Marine Biology and Aquaculture, College of Marine Science, Gyeongsang National University, Tongyeong, South Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, Busan, South Korea
| |
Collapse
|
27
|
Casero D, Gill K, Sridharan V, Koturbash I, Nelson G, Hauer-Jensen M, Boerma M, Braun J, Cheema AK. Space-type radiation induces multimodal responses in the mouse gut microbiome and metabolome. MICROBIOME 2017; 5:105. [PMID: 28821301 PMCID: PMC5563039 DOI: 10.1186/s40168-017-0325-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 08/08/2017] [Indexed: 05/19/2023]
Abstract
BACKGROUND Space travel is associated with continuous low dose rate exposure to high linear energy transfer (LET) radiation. Pathophysiological manifestations after low dose radiation exposure are strongly influenced by non-cytocidal radiation effects, including changes in the microbiome and host gene expression. Although the importance of the gut microbiome in the maintenance of human health is well established, little is known about the role of radiation in altering the microbiome during deep-space travel. RESULTS Using a mouse model for exposure to high LET radiation, we observed substantial changes in the composition and functional potential of the gut microbiome. These were accompanied by changes in the abundance of multiple metabolites, which were related to the enzymatic activity of the predicted metagenome by means of metabolic network modeling. There was a complex dynamic in microbial and metabolic composition at different radiation doses, suggestive of transient, dose-dependent interactions between microbial ecology and signals from the host's cellular damage repair processes. The observed radiation-induced changes in microbiota diversity and composition were analyzed at the functional level. A constitutive change in activity was found for several pathways dominated by microbiome-specific enzymatic reactions like carbohydrate digestion and absorption and lipopolysaccharide biosynthesis, while the activity in other radiation-responsive pathways like phosphatidylinositol signaling could be linked to dose-dependent changes in the abundance of specific taxa. CONCLUSIONS The implication of microbiome-mediated pathophysiology after low dose ionizing radiation may be an unappreciated biologic hazard of space travel and deserves experimental validation. This study provides a conceptual and analytical basis of further investigations to increase our understanding of the chronic effects of space radiation on human health, and points to potential new targets for intervention in adverse radiation effects.
Collapse
Affiliation(s)
- David Casero
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kirandeep Gill
- Department of Oncology, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Gregory Nelson
- Department of Radiation Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Jonathan Braun
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center, Washington DC, 20057, USA.
- Department of Biochemistry and Molecular and & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA.
- GCD-7N Pre-Clinical Science Building, 3900 Reservoir Road NW, Washington DC, 20057, USA.
| |
Collapse
|
28
|
Wang JZ, Du WT, Xu YL, Cheng SZ, Liu ZJ. Gut microbiome-based medical methodologies for early-stage disease prevention. Microb Pathog 2017; 105:122-130. [PMID: 28219830 DOI: 10.1016/j.micpath.2017.02.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/17/2022]
|
29
|
Hudgins LC, Hugo JL, Enayat S, Parker TS, Artis AS, Levine DM. Young, healthy South Asians have enhanced lipogenic sensitivity to dietary sugar. Clin Endocrinol (Oxf) 2017; 86:361-366. [PMID: 27988942 DOI: 10.1111/cen.13293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/16/2016] [Accepted: 12/11/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE South Asians have higher rates of type 2 diabetes and cardiovascular disease compared to most other racial/ethnic groups. Increased hepatic de novo lipogenesis (DNL) in response to dietary sugar may accelerate the development of these chronic diseases in this population. STUDY DESIGN Hepatic DNL in response to a calorically sweetened beverage was measured in an outpatient setting in 15 South Asians and 15 Caucasians with similar and normal body mass indexes, waist circumferences, glucose tolerance and lipid profiles. Blood was sampled before and hourly for 4 h after the ingestion of a single beverage made with glucose (1·5 g/kg) and fructose (1·5 g/kg). The main outcome, DNL, was measured as the increase in %palmitate (16:0) in very low-density lipoprotein (VLDL) triglyceride (TG) over 4 h. RESULTS After the sugar dose, the increase in %16:0 in VLDL TG was significantly greater in South Asians vs Caucasians (P = 0·01). VLDL and total TG also increased to a significantly greater extent in South Asians (P = 0·04 and <0·001, respectively). Although the fasting and postsugar levels of insulin and glucose did not differ between groups, the DNL response significantly correlated with the insulin response to sugar in South Asians (r = 0·56, P = 0·03). CONCLUSIONS Hepatic DNL in response to a sugar challenge was greater in healthy, young South Asians compared to Caucasians despite normal indices of insulin sensitivity, and it correlated with the insulin response. These findings suggest an early, insulin-related, gene-nutrient interaction contributing to the high prevalence of diabetes and coronary disease in this population.
Collapse
Affiliation(s)
- Lisa C Hudgins
- The Rogosin Institute, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Samim Enayat
- Weill Cornell Medical College, New York, NY, USA
| | - Thomas S Parker
- The Rogosin Institute, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | | | - Daniel M Levine
- The Rogosin Institute, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
30
|
Uauy R, Suri DJ, Ghosh S, Kurpad A, Rosenberg IH. Low Circulating Amino Acids and Protein Quality: An Interesting Piece in the Puzzle of Early Childhood Stunting. EBioMedicine 2016; 8:28-29. [PMID: 27428415 PMCID: PMC4919554 DOI: 10.1016/j.ebiom.2016.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 05/19/2016] [Accepted: 05/20/2016] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ricardo Uauy
- London School of Hygiene and Tropical Medicine, London, UK; Nevin Scrimshaw International Nutrition Foundation, Boston, MA, USA
| | | | - Shibani Ghosh
- Nevin Scrimshaw International Nutrition Foundation, Boston, MA, USA; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | | | - Irwin H Rosenberg
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| |
Collapse
|