101
|
Hosangadi D, Smith PG, Giersing BK. Considerations for using ETEC and Shigella disease burden estimates to guide vaccine development strategy. Vaccine 2019; 37:7372-7380. [PMID: 29031690 PMCID: PMC6892262 DOI: 10.1016/j.vaccine.2017.09.083] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/26/2017] [Indexed: 01/09/2023]
Abstract
Enterotoxigenic E. coli (ETEC) and Shigella are enteropathogens causing significant global morbidity and mortality, particularly in low-income countries. No licensed vaccine exists for either pathogen, but candidates are in development, with the most advanced candidates potentially approaching pivotal efficacy testing within the next few years. A positive policy recommendation for introduction of any vaccine, following licensure, depends on evidence of vaccine cost-effectiveness and impact on morbidity and mortality. The mortality estimates for these two pathogens have fluctuated over recent years, which has led to uncertainty in the assessment of their relative public health importance for use in low and middle-income countries. This paper summarizes the various ETEC and Shigella disease burden estimates, based on a review of current literature and informal consultations with leading stakeholders in enteric disease modelling. We discuss the factors that underpin the variability, including differences in the modelling methodology; diagnostic tools used to ascertain diarrheal etiology; epidemiological setting; the data that are available to incorporate; and absolute changes in the total number of diarrheal deaths over time. We consider the further work that will strengthen the evidence needed to support future decision making with respect to recommendations on the relative utility of these vaccines.
Collapse
Affiliation(s)
- Divya Hosangadi
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Peter G Smith
- MRC Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - Birgitte K Giersing
- Initiative for Vaccine Research, World Health Organization, CH-1211 Geneva 27, Switzerland.
| |
Collapse
|
102
|
Hosangadi D, Smith PG, Kaslow DC, Giersing BK. WHO consultation on ETEC and Shigella burden of disease, Geneva, 6–7th April 2017: Meeting report. Vaccine 2019; 37:7381-7390. [DOI: 10.1016/j.vaccine.2017.10.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/03/2017] [Indexed: 10/18/2022]
|
103
|
Ruszkiewicz JA, Tinkov AA, Skalny AV, Siokas V, Dardiotis E, Tsatsakis A, Bowman AB, da Rocha JBT, Aschner M. Brain diseases in changing climate. ENVIRONMENTAL RESEARCH 2019; 177:108637. [PMID: 31416010 PMCID: PMC6717544 DOI: 10.1016/j.envres.2019.108637] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 05/12/2023]
Abstract
Climate change is one of the biggest and most urgent challenges for the 21st century. Rising average temperatures and ocean levels, altered precipitation patterns and increased occurrence of extreme weather events affect not only the global landscape and ecosystem, but also human health. Multiple environmental factors influence the onset and severity of human diseases and changing climate may have a great impact on these factors. Climate shifts disrupt the quantity and quality of water, increase environmental pollution, change the distribution of pathogens and severely impacts food production - all of which are important regarding public health. This paper focuses on brain health and provides an overview of climate change impacts on risk factors specific to brain diseases and disorders. We also discuss emerging hazards in brain health due to mitigation and adaptation strategies in response to climate changes.
Collapse
Affiliation(s)
- Joanna A Ruszkiewicz
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia
| | - Anatoly V Skalny
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, Russia; Trace Element Institute for UNESCO, Lyon, France
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States
| | - João B T da Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
104
|
Kumar A, Allison A, Henry M, Scales A, Fouladkhah AC. Development of Salmonellosis as Affected by Bioactive Food Compounds. Microorganisms 2019; 7:microorganisms7090364. [PMID: 31540475 PMCID: PMC6780870 DOI: 10.3390/microorganisms7090364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/15/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Infections caused by Salmonella serovars are the leading cause of foodborne hospitalizations and deaths in Americans, extensively prevalent worldwide, and pose a considerable financial burden on public health infrastructure and private manufacturing. While a comprehensive review is lacking for delineating the role of dietary components on prevention of Salmonellosis, evidence for the role of diet for preventing the infection and management of Salmonellosis symptoms is increasing. The current study is an evaluation of preclinical and clinical studies and their underlying mechanisms to elaborate the efficacy of bioactive dietary components for augmenting the prevention of Salmonella infection. Studies investigating dietary components such as fibers, fatty acids, amino acids, vitamins, minerals, phenolic compounds, and probiotics exhibited efficacy of dietary compounds against Salmonellosis through manipulation of host bile acids, mucin, epithelial barrier, innate and adaptive immunity and gut microbiota as well as impacting the cellular signaling cascades of the pathogen. Pre-clinical studies investigating synergism and/or antagonistic activities of various bioactive compounds, additional randomized clinical trials, if not curtailed by lack of equipoise and ethical concerns, and well-planned epidemiological studies could augment the development of a validated and evidence-based guideline for mitigating the public health burden of human Salmonellosis through dietary compounds.
Collapse
Affiliation(s)
- Ajay Kumar
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Abimbola Allison
- Public Health Microbiology Laboratory, Tennessee State University, Nashville, TN 37209, USA.
| | - Monica Henry
- Public Health Microbiology Laboratory, Tennessee State University, Nashville, TN 37209, USA.
| | - Anita Scales
- Public Health Microbiology Laboratory, Tennessee State University, Nashville, TN 37209, USA.
| | - Aliyar Cyrus Fouladkhah
- Public Health Microbiology Laboratory, Tennessee State University, Nashville, TN 37209, USA.
- Cooperative Extension Program, Tennessee State University, Nashville, TN 37209, USA.
| |
Collapse
|
105
|
Child-Sensitive WASH Composite Score and the Nutritional Status in Cambodian Children. Nutrients 2019; 11:nu11092142. [PMID: 31500268 PMCID: PMC6770572 DOI: 10.3390/nu11092142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/09/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
Progress in health has occurred in the past decades in Cambodia, in terms of health service access and interventions, but several indicators, including the prevalence of malnourished children, remain alarming. The causes of undernutrition are often linked to inadequate access to water, sanitation and hygiene services but limited evidence exists on the direct association between poor WASH practices and children's' nutritional statuses. This study investigates the relationship between water, sanitation and hygiene practices, defined as the child-sensitive composite score, and the nutritional status of children under five years old, measured as the weight-for-height z-score, mid-upper arm circumference or height-for-age z-score in six districts of Cambodia. The analysis used data from a longitudinal study, comprising extensive data collection on anthropometry, health, nutrition, WASH, and cognitive development. Chronological trends in wasting and stunting were described cross-sectionally, whereas the effect of WASH practices on the nutritional status of children over up to three consecutive study visits was examined with a linear mixed-effects model. The prevalence of wasting decreased during the study while stunting prevalence increased. A small, but significant, association was found between the WASH child-sensitive composite scores and the wasting child anthropometry indicators: weight-for-height z-score or mid-upper arm circumference. Evidence for an association with height-for-age z-score, detecting stunted children, was found when the independent variable was quantified according to global, but not national, guidelines. This study reinforces discordant existing evidence towards a direct association between WASH practices and children's nutritional status, suggesting the need to align nutrition and WASH programmes.
Collapse
|
106
|
Vlasova AN, Takanashi S, Miyazaki A, Rajashekara G, Saif LJ. How the gut microbiome regulates host immune responses to viral vaccines. Curr Opin Virol 2019; 37:16-25. [PMID: 31163292 PMCID: PMC6863389 DOI: 10.1016/j.coviro.2019.05.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
The co-evolution of the microbiota and immune system has forged a mutually beneficial relationship. This relationship allows the host to maintain the balance between active immunity to pathogens and vaccines and tolerance to self-antigens and food antigens. In children living in low-income and middle-income countries, undernourishment and repetitive gastrointestinal infections are associated with the failure of oral vaccines. Intestinal dysbiosis associated with these environmental influences, as well as some host-related factors, compromises immune responses and negatively impacts vaccine efficacy. To understand how immune responses to viral vaccines can be optimally modulated, mechanistic studies of the relationship between the microbiome, host genetics, viral infections and the development and function of the immune system are needed. We discuss the potential role of the microbiome in modulating vaccine responses in the context of a growing understanding of the relationship between the gastrointestinal microbiota, host related factors (including histo-blood group antigens) and resident immune cell populations.
Collapse
Affiliation(s)
- Anastasia N Vlasova
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA.
| | - Sayaka Takanashi
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA; Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ayako Miyazaki
- Division of Viral Disease and Epidemiology, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki 305-0856, Japan
| | - Gireesh Rajashekara
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J Saif
- Food Animal Health Research Program, CFAES, Ohio Agricultural Research and Development Center, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA.
| |
Collapse
|
107
|
Khalil I, Troeger CE, Blacker BF, Reiner RC. Capturing the true burden of Shigella and ETEC: The way forward. Vaccine 2019; 37:4784-4786. [DOI: 10.1016/j.vaccine.2019.01.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/19/2018] [Accepted: 01/16/2019] [Indexed: 01/14/2023]
|
108
|
Benninghoff B, Pereira P, Vetter V. Role of healthcare practitioners in rotavirus disease awareness and vaccination - insights from a survey among caregivers. Hum Vaccin Immunother 2019; 16:138-147. [PMID: 31210567 PMCID: PMC7012184 DOI: 10.1080/21645515.2019.1632685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An online survey was designed to assess awareness and understanding of Rotavirus (RV) gastroenteritis (RVGE), and knowledge and attitudes towards RV vaccination in Germany, Poland, Turkey, Indonesia, the Philippines and Thailand. Survey participants (n = 1500) comprised parents, expectant parents and guardians of children ≤5 years of age who have sole or joint responsibility for health and well-being decisions relating to their child, who were recruited from an online panel and provided their consent for study participation. Participants from most countries had a high level of awareness of RV infections (mean: 82%) and of those aware of RV, a mean of 61% participants were aware that RV was the most common cause of GE, however the majority (mean: 59%) were unaware that nearly every child would be infected with RVGE by the age of 5 years. Healthcare professional (HCP) recommendation was identified as the key driver for participants seeking vaccination (48%–75% of participants stated this reason, with results differing by country) followed by availability of RV vaccine in the national immunization program. Despite a high level of awareness of RVGE among participants, fostering knowledge regarding the difficulty of RVGE prevention, the risk of RV contraction and the associated serious consequences like dehydration is imperative to improve RV vaccination uptake. HCPs, being the primary influence on participants’ decision on vaccination, are best suited to bridge existing knowledge gaps and recommend parents to vaccinate their children against RVGE.
Collapse
|
109
|
Grenov B, Lanyero B, Nabukeera-Barungi N, Namusoke H, Ritz C, Friis H, Michaelsen KF, Mølgaard C. Diarrhea, Dehydration, and the Associated Mortality in Children with Complicated Severe Acute Malnutrition: A Prospective Cohort Study in Uganda. J Pediatr 2019; 210:26-33.e3. [PMID: 30992218 DOI: 10.1016/j.jpeds.2019.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To assess predictors of diarrhea and dehydration and to investigate the role of diarrhea in mortality among children with complicated severe acute malnutrition. STUDY DESIGN A prospective cohort study, nested in a probiotic trial, was conducted in children with complicated severe acute malnutrition. Children were treated according to World Health Organization and national guidelines, and diarrhea and dehydration were assessed daily. Multiple linear and log-linear Poisson regression models were used to identify predictors of days with diarrhea and dehydration, respectively, and multiple logistic regression was used to assess their role in mortality. RESULTS Among 400 children enrolled, the median (IQR) age was 15.0 months (11.2-19.2 months), 58% were boys, and 61% had caregiver-reported diarrhea at admission. During hospitalization, the median (range) number of days with diarrhea was 5 (0-31), the median duration of hospitalization was 17 days (1-69 days), and 39 (10%) died. Of 592 diarrhea episodes monitored, 237 were admission episodes and 355 were hospital acquired. During hospitalization, young age was associated with days with diarrhea, and young age and HIV infection were associated with dehydration. Both days with diarrhea and dehydration predicted duration of hospitalization as well as mortality. The odds of mortality increased by a factor of 1.4 (95% CI, 1.2-1.6) per day of diarrhea and 3.5 (95% CI, 2.2-6.0) per unit increase in dehydration score. CONCLUSIONS Diarrhea is a strong predictor of mortality among children with complicated severe acute malnutrition. Improved management of diarrhea and prevention of hospital-acquired diarrhea may be critical to decreasing mortality.
Collapse
Affiliation(s)
- Benedikte Grenov
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark.
| | - Betty Lanyero
- Mwanamugimu Nutrition Unit, Department of Pediatrics and Child Health, Mulago National Referral Hospital, Kampala, Uganda
| | | | - Hanifa Namusoke
- Mwanamugimu Nutrition Unit, Department of Pediatrics and Child Health, Mulago National Referral Hospital, Kampala, Uganda
| | - Christian Ritz
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Friis
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
110
|
DeBoer MD, Guerrant RL. Ongoing Challenges to Understanding and Interrupting Environmental Enteric Dysfunction. J Pediatr 2019; 210:8-9. [PMID: 30926153 DOI: 10.1016/j.jpeds.2019.02.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 11/20/2022]
Affiliation(s)
- Mark D DeBoer
- Pediatric Endocrinology, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Infectious Diseases and International Health, Department of Medicine and, Center for Global Health, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
111
|
Malhotra I, LaBeaud AD, Morris N, McKibben M, Mungai P, Muchiri E, King CL, King CH. Cord Blood Antiparasite Interleukin 10 as a Risk Marker for Compromised Vaccine Immunogenicity in Early Childhood. J Infect Dis 2019; 217:1426-1434. [PMID: 29390149 DOI: 10.1093/infdis/jiy047] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 01/23/2018] [Indexed: 01/09/2023] Open
Abstract
Background Antenatal exposure to parasites can affect infants' subsequent responses to vaccination. The present study investigated how maternal prenatal infections and newborns' antiparasite cytokine profiles relate to immunoglobulin G (IgG) responses to standard vaccination during infancy. Methods A total of 450 Kenyan women were tested for parasitic infections during pregnancy. Their newborns' responses to Plasmodium falciparum, schistosome, and filaria antigens were assessed in cord blood lymphocytes. Following standard neonatal vaccination, this infant cohort was followed biannually to age 30 months for measurement of circulating IgG levels against Haemophilus influenzae b (Hib), diphtheria toxoid (DT), hepatitis B virus (HBV), and tetanus toxoid. Results Trajectories of postvaccination IgG levels were classified by functional principal component (PC) analysis to assess each child's response profile. Two main components, PC1, reflecting height of response over time, and PC2, reflecting crossover from high to low responses or from low to high responses, were identified. Cord blood cytokine responses to schistosome and filarial antigens showed a significant association between augmented antihelminth interleukin 10 and reduced antibody levels, particularly to DT and HBV, and a more rapid postvaccination decline in circulating IgG levels against Hib. Conclusion Antenatal sensitization to schistosomiasis or filariasis and related production of antiparasite interleukin 10 at birth are associated with reduced antivaccine IgG levels in infancy, with possibly impaired protection.
Collapse
Affiliation(s)
- Indu Malhotra
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio.,Clinical and Translational Science Collaborative, Case Western Reserve University, Cleveland, Ohio
| | - A Desiree LaBeaud
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio.,Child Health Research Institute, Stanford, California
| | - Nathan Morris
- Clinical and Translational Science Collaborative, Case Western Reserve University, Cleveland, Ohio
| | - Maxim McKibben
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| | - Peter Mungai
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Eric Muchiri
- Division of Vector Borne and Neglected Tropical Diseases, Ministry of Public Health and Sanitation, Nairobi, Kenya
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| | - Charles H King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
112
|
Srugo SA, Bloise E, Nguyen TTTN, Connor KL. Impact of Maternal Malnutrition on Gut Barrier Defense: Implications for Pregnancy Health and Fetal Development. Nutrients 2019; 11:nu11061375. [PMID: 31248104 PMCID: PMC6628366 DOI: 10.3390/nu11061375] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
Small intestinal Paneth cells, enteric glial cells (EGC), and goblet cells maintain gut mucosal integrity, homeostasis, and influence host physiology locally and through the gut-brain axis. Little is known about their roles during pregnancy, or how maternal malnutrition impacts these cells and their development. Pregnant mice were fed a control diet (CON), undernourished by 30% vs. control (UN), or fed a high fat diet (HF). At day 18.5 (term = 19), gut integrity and function were assessed by immunohistochemistry and qPCR. UN mothers displayed reduced mRNA expression of Paneth cell antimicrobial peptides (AMP; Lyz2, Reg3g) and an accumulation of villi goblet cells, while HF had reduced Reg3g and mucin (Muc2) mRNA and increased lysozyme protein. UN fetuses had increased mRNA expression of gut transcription factor Sox9, associated with reduced expression of maturation markers (Cdx2, Muc2), and increased expression of tight junctions (TJ; Cldn-7). HF fetuses had increased mRNA expression of EGC markers (S100b, Bfabp, Plp1), AMP (Lyz1, Defa1, Reg3g), and TJ (Cldn-3, Cldn-7), and reduced expression of an AMP-activator (Tlr4). Maternal malnutrition altered expression of genes that maintain maternal gut homeostasis, and altered fetal gut permeability, function, and development. This may have long-term implications for host-microbe interactions, immunity, and offspring gut-brain axis function.
Collapse
Affiliation(s)
- Sebastian A Srugo
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Enrrico Bloise
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil.
| | | | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
113
|
Alvestegui A, Olivares-Morales M, Muñoz E, Smith R, Nataro JP, Ruiz-Perez F, Farfan MJ. TLR4 Participates in the Inflammatory Response Induced by the AAF/II Fimbriae From Enteroaggregative Escherichia coli on Intestinal Epithelial Cells. Front Cell Infect Microbiol 2019; 9:143. [PMID: 31131263 PMCID: PMC6509964 DOI: 10.3389/fcimb.2019.00143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/17/2019] [Indexed: 12/21/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) infections are one of the most frequent causes of persistent diarrhea in children, immunocompromised patients and travelers worldwide. The most prominent colonization factors of EAEC are aggregative adherence fimbriae (AAF). EAEC prototypical strain 042 harbors the AAF/II fimbriae variant, which mediates adhesion to intestinal epithelial cells and participates in the induction of an inflammatory response against this pathogen. However, the mechanism and the cell receptors implicated in eliciting this response have not been fully characterized. Since previous reports have shown that TLR4 recognize fimbriae from different pathogens, we evaluated the role of this receptor in the response elicited against EAEC by intestinal cells. Using a mutual antagonist against TLR2 and TLR4 (OxPAPC), we observed that blocking of these receptors significantly reduces the secretion of the inflammatory marker IL-8 in response to EAEC and AAF/II fimbrial extract in HT-29 cells. Using a TLR4-specific antagonist (TAK-242), we observed that the secretion of this cytokine was significantly reduced in HT-29 cells infected with EAEC or incubated with AAF/II fimbrial extract. We evaluated the participation of AAF/II fimbriae in the TLR4-mediated secretion of 38 cytokines, chemokines, and growth factors involved in inflammation. A reduction in the secretion of IL-8, GRO, and IL-4 was observed. Our results suggest that TLR4 participates in the secretion of several inflammation biomarkers in response to AAF/II fimbriae.
Collapse
Affiliation(s)
- Alejandra Alvestegui
- Departamento de Pediatría, Facultad de Medicina, Centro de Estudios Moleculares, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Mauricio Olivares-Morales
- Departamento de Pediatría, Facultad de Medicina, Centro de Estudios Moleculares, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Ernesto Muñoz
- Departamento de Pediatría, Facultad de Medicina, Centro de Estudios Moleculares, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Rachel Smith
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Mauricio J Farfan
- Departamento de Pediatría, Facultad de Medicina, Centro de Estudios Moleculares, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| |
Collapse
|
114
|
Cryptosporidium parvum Subtilisin-Like Serine Protease (SUB1) Is Crucial for Parasite Egress from Host Cells. Infect Immun 2019; 87:IAI.00784-18. [PMID: 30782859 DOI: 10.1128/iai.00784-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/28/2019] [Indexed: 01/28/2023] Open
Abstract
Despite the severity and global burden of Cryptosporidium infection, treatments are less than optimal, and there is no effective vaccine. Egress from host cells is a key process for the completion of the life cycle of apicomplexan parasites. For Plasmodium species, subtilisin-like serine protease (SUB1) is a key mediator of egress. For Toxoplasma species, calcium-dependent protein kinases (CDPKs) are critical. In this study, we characterized Cryptosporidium SUB1 expression and evaluated its effect using an infection model. We found increased expression between 12 and 20 h after in vitro infection, prior to egress. We induced silencing of SUB1 (ΔSUB1) mRNA using SUB1 single-stranded antisense RNA coupled with human Argonaute 2. Silencing of SUB1 mRNA expression did not affect parasite viability, excystation, or invasion of target cells. However, knockdown led to a 95% decrease in the proportion of released merozoites in vitro (P < 0.0001). In contrast, silencing of CDPK5 had no effect on egress. Overall, our results indicate that SUB1 is a key mediator of Cryptosporidium egress and suggest that interruption of the life cycle at this stage may effectively inhibit the propagation of infection.
Collapse
|
115
|
Jesson J, Ephoevi-Ga A, Desmonde S, Ake-Assi MH, D'Almeida M, Sy HS, Malateste K, Amorissani-Folquet M, Dicko F, Kouadio K, Renner L, Leroy V. Growth in the first 5 years after antiretroviral therapy initiation among HIV-infected children in the IeDEA West African Pediatric Cohort. Trop Med Int Health 2019; 24:775-785. [PMID: 30945378 DOI: 10.1111/tmi.13237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To describe growth evolution and its correlates in the first 5 years of antiretroviral therapy (ART) initiation among HIV-infected children followed up in West Africa. METHODS All HIV-infected children younger than 10 years followed in the IeDEA pWADA cohort while initiating ART, with at least one anthropometric measurement within the first 5 years of treatment were included in the study. Growth was described according to the WHO child growth standards, using Weight-for-age Z-score (WAZ), Height-for-age Z-score (HAZ) and Weight-for-Height/BMI-for-age Z-score (WHZ/BAZ). Growth evolution and its correlates, measured at ART initiation, were modelled in individual linear mixed models for each anthropometric indicator, with a spline term added at the 12-, 24- and 9-month time point for WAZ, HAZ and WHZ/BAZ, respectively. RESULTS Among the 4156 children selected (45% girls, median age at ART initiation 3.9 years [IQR interquartile range 1.9-6.6], and overall 68% malnourished at ART initiation), important gains were observed in the first 12, 24 and 9 months on ART for WAZ, HAZ and WHZ/BAZ, respectively. Correlates at ART initiation of a better growth evolution overtime were early age (<2 years of age), severe immunodeficiency for age, and severity of malnutrition. CONCLUSIONS Growth evolution is particularly strong within the first 2 years on ART but slows down after this period. Weight and height gains help to recover from pre-ART growth deficiency but are insufficient for the most severely malnourished. The first year on ART could be the best period for nutritional interventions to optimize growth among HIV-infected children in the long-term.
Collapse
Affiliation(s)
- Julie Jesson
- Inserm U1027, Université Paul Sabatier Toulouse 3, Toulouse, France
| | | | - Sophie Desmonde
- Inserm U1027, Université Paul Sabatier Toulouse 3, Toulouse, France
| | | | | | | | - Karen Malateste
- Inserm U1219, Bordeaux Population Health Center, Université de Bordeaux, Bordeaux, France
| | | | | | | | | | - Valériane Leroy
- Inserm U1027, Université Paul Sabatier Toulouse 3, Toulouse, France
| | | |
Collapse
|
116
|
Liu H, Guo M, Jiang Y, Cao Y, Qian Q, He X, Huang K, Zhang J, Xu W. Diagnosing and tracing the pathogens of infantile infectious diarrhea by amplicon sequencing. Gut Pathog 2019; 11:12. [PMID: 30992716 PMCID: PMC6451272 DOI: 10.1186/s13099-019-0292-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/19/2019] [Indexed: 01/14/2023] Open
Abstract
Background Metagenomic methods have been widely applied to study the relationship between gut microbiota and human health. To test whether metagenomic amplicon sequencing could be an effective method to diagnose and trace the pathogens of infantile infectious diarrhea, the fecal samples of 20 diarrheic and 13 healthy infants were collected. After 16S rDNA amplicon sequencing, diversity analyses were carried out. The relationship between the pathogens of the gut microbiota and geography of patients was analyzed. Results The diversity of the gut microbiota in diarrheic infants was significantly lower than that of the gut microbiota in healthy ones and that, the composition of gut microbiota in the diarrheic group was significantly different than that of the gut microbiota in the healthy group. The results also indicated that in some of the patients, the amounts of Escherichia coli were significantly increased in the diarrheic infants, which was in agreement with the result of the qPCR analysis. Using a geographical map, we found some patterns between pathogen source and geographical location. This is helpful for an early warning of the disease. Conclusions The method of using high-throughput DNA sequencing and a comprehensive and deep data analysis can be a new strategy to detect and trace pathogens in infantile infectious diarrhea. Trial registration Diagnosing and tracing the pathogens of infantile infectious diarrhea by amplicon sequencing, ChiCTR-DDD-1701088, Registered 16 March 2017-Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=18477
Collapse
Affiliation(s)
- Haiyan Liu
- 1School of Public Health, North China University of Science and Technology, Tangshan, Hebei China
| | - Mingzhang Guo
- 2Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yuanchunzi Jiang
- 2Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanhua Cao
- 1School of Public Health, North China University of Science and Technology, Tangshan, Hebei China
| | - Qingzeng Qian
- 1School of Public Health, North China University of Science and Technology, Tangshan, Hebei China
| | - Xiaoyun He
- 2Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kunlun Huang
- 2Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jianwei Zhang
- 3Physical Education Department, Tangshan Normal University, Tangshan, Hebei China
| | - Wentao Xu
- 2Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China.,4Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Haidian District, Beijing, 100083 China
| |
Collapse
|
117
|
Blaney S, Menasria L, Main B, Chhorvann C, Vong L, Chiasson L, Hun V, Raminashvili D. Determinants of Undernutrition among Young Children Living in Soth Nikum District, Siem Reap, Cambodia. Nutrients 2019; 11:E685. [PMID: 30909463 PMCID: PMC6471553 DOI: 10.3390/nu11030685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Child undernutrition is of public concern in Cambodia. An understanding of factors influencing child nutritional status is essential to design programs that will reduce undernutrition. Using the UNICEF conceptual framework of causes of malnutrition, our research investigates the relationship between nutritional status of children aged 6⁻23 months and its immediate and underlying determinants. METHODS Baseline data from a cluster-randomized controlled trial aiming to assess the impact of the promotion of optimal feeding practices combined or not with the provision of local foods among 360 children 6⁻23 months of age were used. Anthropometry and biochemical measurements were performed at baseline. Data on each determinant of undernutrition were collected through interviews and direct observations. RESULTS Our results show that the degree of satisfaction of proteins and zinc requirements as well as the access to improved water sources and sanitation were positively associated with length-for-age, while having a better health status and a higher degree of satisfaction of energy, protein, zinc, and iron requirements were associated to an improved weight-for-length. Only child health status was associated to ferritin. CONCLUSION Our results reiterate the importance of improving child diet and health status, but also the access to a healthy environment to ensure an optimal nutritional status.
Collapse
Affiliation(s)
- Sonia Blaney
- École des sciences des aliments, de nutrition et d'étude familiale, Université de Moncton, 18 avenue Antonine-Maillet, Moncton, NB E1A 3E9, Canada.
| | - Lylia Menasria
- École des sciences des aliments, de nutrition et d'étude familiale, Université de Moncton, 18 avenue Antonine-Maillet, Moncton, NB E1A 3E9, Canada.
| | - Barbara Main
- Public Health Specialist, Guelph, ON N1E 6Y8, Canada.
| | - Chhea Chhorvann
- National Institute of Public Health, Phnom Penh 12203, Cambodia.
| | - Lenin Vong
- Independent consultant, Phnom Penh 12203, Cambodia.
| | - Lucie Chiasson
- Direction du mieux-être, Ministère du développement social, 1780 rue Water, Miramichi, NB E1N 1B6, Canada.
| | - Vannary Hun
- World Vision Cambodia, #20 Street 71 Tonle Bassac, Chamkar Morn, Phnom Penh 12203, Cambodia.
| | - David Raminashvili
- World Vision Cambodia, #20 Street 71 Tonle Bassac, Chamkar Morn, Phnom Penh 12203, Cambodia.
| |
Collapse
|
118
|
Gladstone MJ, Chandna J, Kandawasvika G, Ntozini R, Majo FD, Tavengwa NV, Mbuya MNN, Mangwadu GT, Chigumira A, Chasokela CM, Moulton LH, Stoltzfus RJ, Humphrey JH, Prendergast AJ. Independent and combined effects of improved water, sanitation, and hygiene (WASH) and improved complementary feeding on early neurodevelopment among children born to HIV-negative mothers in rural Zimbabwe: Substudy of a cluster-randomized trial. PLoS Med 2019; 16:e1002766. [PMID: 30897095 PMCID: PMC6428259 DOI: 10.1371/journal.pmed.1002766] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/21/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Globally, nearly 250 million children (43% of all children under 5 years of age) are at risk of compromised neurodevelopment due to poverty, stunting, and lack of stimulation. We tested the independent and combined effects of improved water, sanitation, and hygiene (WASH) and improved infant and young child feeding (IYCF) on early child development (ECD) among children enrolled in the Sanitation Hygiene Infant Nutrition Efficacy (SHINE) trial in rural Zimbabwe. METHODS AND FINDINGS SHINE was a cluster-randomized community-based 2×2 factorial trial. A total of 5,280 pregnant women were enrolled from 211 clusters (defined as the catchment area of 1-4 village health workers [VHWs] employed by the Zimbabwean Ministry of Health and Child Care). Clusters were randomly allocated to standard of care, IYCF (20 g of small-quantity lipid-based nutrient supplement per day from age 6 to 18 months plus complementary feeding counseling), WASH (ventilated improved pit latrine, handwashing stations, chlorine, liquid soap, and play yard), and WASH + IYCF. Primary outcomes were child length-for-age Z-score and hemoglobin concentration at 18 months of age. Children who completed the 18-month visit and turned 2 years (102-112 weeks) between March 1, 2016, and April 30, 2017, were eligible for the ECD substudy. We prespecified that primary inferences would be drawn from findings of children born to HIV-negative mothers; these results are presented in this paper. A total of 1,655 HIV-unexposed children (64% of those eligible) were recruited into the ECD substudy from 206 clusters and evaluated for ECD at 2 years of age using the Malawi Developmental Assessment Tool (MDAT) to assess gross motor, fine motor, language, and social skills; the MacArthur-Bates Communicative Development Inventories (CDI) to assess vocabulary and grammar; the A-not-B test to assess object permanence; and a self-control task. Outcomes were analyzed in the intention-to-treat population. For all ECD outcomes, there was not a statistical interaction between the IYCF and WASH interventions, so we estimated the effects of the interventions by comparing the 2 IYCF groups with the 2 non-IYCF groups and the 2 WASH groups with the 2 non-WASH groups. The mean (95% CI) total MDAT score was modestly higher in the IYCF groups compared to the non-IYCF groups in unadjusted analysis: 1.35 (0.24, 2.46; p = 0.017); this difference did not persist in adjusted analysis: 0.79 (-0.22, 1.68; p = 0.057). There was no evidence of impact of the IYCF intervention on the CDI, A-not-B, or self-control tests. Among children in the WASH groups compared to those in the non-WASH groups, mean scores were not different for the MDAT, A-not-B, or self-control tests; mean CDI score was not different in unadjusted analysis (0.99 [95% CI -1.18, 3.17]) but was higher in children in the WASH groups in adjusted analysis (1.81 [0.01, 3.61]). The main limitation of the study was the specific time window for substudy recruitment, meaning not all children from the main trial were enrolled. CONCLUSIONS We found little evidence that the IYCF and WASH interventions implemented in SHINE caused clinically important improvements in child development at 2 years of age. Interventions that directly target neurodevelopment (e.g., early stimulation) or that more comprehensively address the multifactorial nature of neurodevelopment may be required to support healthy development of vulnerable children. TRIAL REGISTRATION ClinicalTrials.gov NCT01824940.
Collapse
Affiliation(s)
- Melissa J. Gladstone
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| | - Jaya Chandna
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D. Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V. Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mduduzi N. N. Mbuya
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Global Alliance for Improved Nutrition, Washington, District of Columbia, United States of America
| | | | | | | | - Lawrence H. Moulton
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Rebecca J. Stoltzfus
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
| | - Jean H. Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Andrew J. Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
119
|
Tsai K, Simiyu S, Mumma J, Aseyo RE, Cumming O, Dreibelbis R, Baker KK. Enteric Pathogen Diversity in Infant Foods in Low-Income Neighborhoods of Kisumu, Kenya. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16030506. [PMID: 30759722 PMCID: PMC6388216 DOI: 10.3390/ijerph16030506] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 01/01/2023]
Abstract
Pediatric diarrheal disease remains the second most common cause of preventable illness and death among children under the age of five, especially in low and middle-income countries (LMICs). However, there is limited information regarding the role of food in pathogen transmission in LMICs. For this study, we examined the frequency of enteric pathogen occurrence and co-occurrence in 127 infant weaning foods in Kisumu, Kenya, using a multi-pathogen PCR diagnostic tool, and assessed household food hygiene risk factors for contamination. Bacterial, viral, and protozoan enteric pathogen DNA and RNA were detected in 62% of the infant weaning food samples collected, with 37% of foods containing more than one pathogen type. Multivariable generalized linear mixed model analysis indicated type of infant food best explained the presence and diversity of enteric pathogens in infant food, while most household food hygiene risk factors considered in this study were not significantly associated with pathogen contamination. Specifically, cow’s milk was significantly more likely to contain a pathogen (adjusted risk ratio = 14.4; 95% confidence interval (CI) 1.78–116.1) and more likely to have higher number of enteric pathogen species (adjusted risk ratio = 2.35; 95% CI 1.67–3.29) than porridge. Our study demonstrates that infants in this low-income urban setting are frequently exposed to diarrhoeagenic pathogens in food and suggests that interventions are needed to prevent foodborne transmission of pathogens to infants.
Collapse
Affiliation(s)
- Kevin Tsai
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA.
| | - Sheillah Simiyu
- Center of Research, Great Lakes University of Kisumu, Kisumu 40100, Kenya.
| | - Jane Mumma
- Center of Research, Great Lakes University of Kisumu, Kisumu 40100, Kenya.
| | - Rose Evalyne Aseyo
- Center of Research, Great Lakes University of Kisumu, Kisumu 40100, Kenya.
| | - Oliver Cumming
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK.
| | - Robert Dreibelbis
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK.
| | - Kelly K Baker
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52246, USA.
| |
Collapse
|
120
|
do Carmo MS, Santos CID, Araújo MC, Girón JA, Fernandes ES, Monteiro-Neto V. Probiotics, mechanisms of action, and clinical perspectives for diarrhea management in children. Food Funct 2019; 9:5074-5095. [PMID: 30183037 DOI: 10.1039/c8fo00376a] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Infectious diarrhea is the second most common cause of morbidity and mortality in children under 5 years of age in the underdeveloped areas of the world. Conventional treatment consists of rehydration, which may be coupled with antimicrobial agents in more severe bacterial infections or with antiprotozoal agents. In the last few decades, research on the use of probiotic strains, such as Lactobacillus rhamnosus GG ATCC 53013 (LGG), Lactobacillus reuteri DSM 17938 and Saccharomyces boulardii, has gained much attention to prevent and treat diarrheal diseases. However, they are rarely used in the clinical routine, perhaps because there are still gaps in the knowledge about the effective benefit to the patient in terms of the reduction of the duration of diarrhea and its prevention. Furthermore, only a few probiotic strains are safely indicated for usage in pediatric practice. This review summarizes the current knowledge on the antimicrobial mechanisms of probiotics on distinct enteropathogens and their role in stimulating host defense mechanisms against intestinal infections. In addition, we highlight the potential of probiotics for the treatment and prevention of diarrhea in children. We conclude that the use of probiotics is beneficial for both the treatment and prevention of diarrhea in children and that the identification of other candidate probiotics might represent an important advance to a greater reduction in hospital stays and to prevent infectious diarrhea in a larger portion of this population.
Collapse
Affiliation(s)
- Monique Santos do Carmo
- Programa de Pós-graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís, MA, Brazil
| | | | | | | | | | | |
Collapse
|
121
|
Heat-Stable Enterotoxins of Enterotoxigenic Escherichia coli and Their Impact on Host Immunity. Toxins (Basel) 2019; 11:toxins11010024. [PMID: 30626031 PMCID: PMC6356903 DOI: 10.3390/toxins11010024] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/31/2018] [Accepted: 01/03/2019] [Indexed: 01/12/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are an important diarrhea-causing pathogen and are regarded as a global threat for humans and farm animals. ETEC possess several virulence factors to infect its host, including colonization factors and enterotoxins. Production of heat-stable enterotoxins (STs) by most ETEC plays an essential role in triggering diarrhea and ETEC pathogenesis. In this review, we summarize the heat-stable enterotoxins of ETEC strains from different species as well as the molecular mechanisms used by these heat-stable enterotoxins to trigger diarrhea. As recently described, intestinal epithelial cells are important modulators of the intestinal immune system. Thus, we also discuss the impact of the heat-stable enterotoxins on this role of the intestinal epithelium and how these enterotoxins might affect intestinal immune cells. Finally, the latest developments in vaccination strategies to protect against infections with ST secreting ETEC strains are discussed. This review might inform and guide future research on heat-stable enterotoxins to further unravel their molecular pathogenesis, as well as to accelerate vaccine design.
Collapse
|
122
|
van Sadelhoff JHJ, Perez Pardo P, Wu J, Garssen J, van Bergenhenegouwen J, Hogenkamp A, Hartog A, Kraneveld AD. The Gut-Immune-Brain Axis in Autism Spectrum Disorders; A Focus on Amino Acids. Front Endocrinol (Lausanne) 2019; 10:247. [PMID: 31057483 PMCID: PMC6477881 DOI: 10.3389/fendo.2019.00247] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/29/2019] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a range of neurodevelopmental conditions that affect communication and social behavior. Besides social deficits, systemic inflammation, gastrointestinal immune-related problems, and changes in the gut microbiota composition are characteristic for people with ASD. Animal models showed that these characteristics can induce ASD-associated behavior, suggesting an intimate relationship between the microbiota, gut, immune system and the brain in ASD. Multiple factors can contribute to the development of ASD, but mutations leading to enhanced activation of the mammalian target of rapamycin (mTOR) are reported frequently. Hyperactivation of mTOR leads to deficits in the communication between neurons in the brain and to immune impairments. Hence, mTOR might be a critical factor linking the gut-brain-immune axis in ASD. Pharmacological inhibition of mTOR is shown to improve ASD-associated behavior and immune functions, however, the clinical use is limited due to severe side reactions. Interestingly, studies have shown that mTOR activation can also be modified by nutritional stimuli, in particular by amino acids. Moreover, specific amino acids are demonstrated to inhibit inflammation, improve gut barrier function and to modify the microbiota composition. In this review we will discuss the gut-brain-immune axis in ASD and explore the potential of amino acids as a treatment option for ASD, either via modification of mTOR activity, the immune system or the gut microbiota composition.
Collapse
Affiliation(s)
- Joris H. J. van Sadelhoff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jiangbo Wu
- Laboratory of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anita Hartog
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Veterinary Pharmacology, Institute for Risk Assessment Studies, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
- *Correspondence: Aletta D. Kraneveld
| |
Collapse
|
123
|
Genetic Diversity of Norovirus Infections, Coinfections, and Undernutrition in Children From Brazilian Semiarid Region. J Pediatr Gastroenterol Nutr 2018; 67:e117-e122. [PMID: 29985875 DOI: 10.1097/mpg.0000000000002085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND OBJECTIVE Norovirus (NoV) infections are known to have high-morbidity and mortality rates and are a major health problem globally. The impact of NoV on child development is, however, poorly understood. We evaluated the distribution of NoV genotypes in children from a low-income Brazilian semiarid region, in relation with their clinical symptoms, nutritional status, and co-pathogens. METHODS The test population included children aged 2 to 36 months from 6 cities of the Brazilian semiarid region. Fecal samples were collected from each child, along with the information regarding their socioeconomic/clinical conditions using a standardized questionnaire. Detection and quantification of NoV were performed by reverse-transcription quantitative polymerase chain reaction, followed by molecular and phylogenetic analyses. RESULTS The NoV detection rate was 45.2%. Presence of NoV was associated with lower z scores for weight-for-age (P = 0.03), weight-for-height (P = 0.03), and body mass index-for-age (P = 0.03). NoV infection was associated with more frequent respiratory illnesses (P < 0.01). GII.P7 (polymerase) and GII.3 (capsid) were the most frequent NoV genotypes. Analysis of the open reading frame (ORF)1-2 junction identified recombinant NoV strains in 80% of the sequenced samples. Enteroaggregative Escherichia coli coinfection was the major predictor for diarrhea in NoV-positive samples (P < 0.02). Moreover, Shigella spp was also associated with NoV-positive diagnosis (P = 0.02). CONCLUSIONS This study highlights the genetic variability of NoV and, associated co-infections and undernutrition in infants from low-income Brazilian semiarid region.
Collapse
|
124
|
Abstract
The volume of research into the pathogenesis and treatment of malnutrition has increased markedly over the past ten years, providing mechanistic insights that can be leveraged into more effective treatment options. These discoveries have been driven by several landmark studies employing metabolomics, metagenomics, and new preclinical models. This review highlights some of the most important recent findings, focusing in particular on the emerging roles of prenatal and perinatal factors, protein deficiency, impaired gut barrier function, immune deficiency, and the intestinal microbiome.
Collapse
|
125
|
Risk factors for diarrhoea and malnutrition among children under the age of 5 years in the Tigray Region of Northern Ethiopia. PLoS One 2018; 13:e0207743. [PMID: 30475875 PMCID: PMC6257922 DOI: 10.1371/journal.pone.0207743] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 11/06/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Diarrhoea and malnutrition are the leading cause of morbidity and mortality among children in areas with poor access to clean water, improved sanitation, and with low socioeconomic status. This study was designed to determine the prevalence of diarrhoea, malnutrition and risk factors among children aged 6-59 months in the Tigray Region of Northern Ethiopia. METHODS A community based cross-sectional study design was conducted from June to August 2017 to assess the magnitude and factors associated with diarrhoea and malnutrition among children. A standardized questionnaire was used to collect data on diarrhoea, environmental, demographic and behavioural factors from 610 mother-child pairs. Anthropometric measurements were collected from the children. SPSS ver.21 statistical software was used for analysis. Factors associated with diarrhoea and nutritional status were identified using bivariate and multivariate logistic regression. A p-value ≤ 0.05 was considered statistically significant. RESULTS Of the 610 children monitored in this study, the incidence of diarrhoea among 6-59 month-old children in the two weeks preceding the day of the interview day was 27.2% (95% CI: 23.6-31%). Specifically, 35.9%, 9.7%, and 1.8% had 1-2, 3-4 and 5-6 times of diarrhoea episodes in a one year of time, respectively. The prevalence of stunting, underweight, wasting, and acute under-nutrition were 36.1% (95% CI: 31-38.6%), 37% (95% CI: 32-39.6%), 7.9% (95% CI: 5.5-9.7%), and 5.4% (95% CI: 3.8-7.4%), respectively. In a multivariate logistic regression analysis, type of drinking water source [AOR = 3.69; 95% CI: 2.03-6.71], mothers not hand washing at critical times [AOR = 15.42; 95% CI: 2.02-117.78], improper solid waste disposal [AOR = 12.81; 95% CI: 2.50-65.62], and child age (36-47 months) [AOR = 2.57; 95% CI: 1.45-4.55] were found to be predictors of diarrhoea. Being within the age range of 12-23 months was a predictor for wasting [AOR = 4.38; 95% CI: 1.61-11.90] and being underweight [AOR = 4.4; 95% CI: 1.7-11.2]. Similarly, the age range of 36-47 months was associated with wasting [AOR = 2.3; 95% CI: 1.45-3.85] and stunting [AOR = 1.7; 95% CI: 1.03-2.67]. Family size (less than 4) [AOR = 0.56; 95% CI: 0.368-0.959] was inversely associated for wasting. CONCLUSIONS Our study revealed that the problem of diarrhoea and malnutrition amongst 6-59 months children in the study area was significant. Access to clean water was the main problem in the study area. Hence, improving access to clean water and providing health education to mothers on personal and environmental hygiene, and proper waste disposal could improve diarrhoea in the study area. Intervention on children's nutrition should also be implemented to minimize the problem of malnutrition.
Collapse
|
126
|
Kim MJ, Moon YH, Kim H, Rho S, Shin YK, Song M, Walker R, Czerkinsky C, Kim DW, Kim JO. Cross-Protective Shigella Whole-Cell Vaccine With a Truncated O-Polysaccharide Chain. Front Microbiol 2018; 9:2609. [PMID: 30429838 PMCID: PMC6220597 DOI: 10.3389/fmicb.2018.02609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/12/2018] [Indexed: 11/13/2022] Open
Abstract
Shigella is a highly prevalent bacterium causing acute diarrhea and dysentery in developing countries. Shigella infections are treated with antibiotics but Shigellae are increasingly resistant to these drugs. Vaccination can be a countermeasure against emerging antibiotic-resistant shigellosis. Because of the structural variability in Shigellae O-antigen polysaccharides (Oag), cross-protective Shigella vaccines cannot be derived from single serotype-specific Oag. We created an attenuated Shigella flexneri 2a strain with one rather than multiple Oag units by disrupting the Oag polymerase gene (Δwzy), which broadened protective immunogenicity by exposing conserved surface proteins. Inactivated Δwzy mutant cells combined with Escherichia coli double mutant LT(R192G/L211A) as adjuvant, induced potent antibody responses to outer membrane protein PSSP-1, and type III secretion system proteins IpaB and IpaC. Intranasal immunization with the vaccine preparation elicited cross-protective immunity against S. flexneri 2a, S. flexneri 3a, S. flexneri 6, and Shigella sonnei in a mouse pneumonia model. Thus, S. flexneri 2a Δwzy represents a promising candidate strain for a universal Shigella vaccine.
Collapse
Affiliation(s)
- Min Jung Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea.,Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Young-Hye Moon
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Heejoo Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Semi Rho
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Manki Song
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | | | - Cecil Czerkinsky
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea.,Institut de Pharmacologie Moléculaire & Cellulaire CNRS-INSERM-University of Nice Sophia Antipolis, Valbonne, France
| | - Dong Wook Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Jae-Ouk Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| |
Collapse
|
127
|
Reducing amount and frequency of meal as a major coping strategy for food insecurity. ACTA ACUST UNITED AC 2018; 76:56. [PMID: 30305899 PMCID: PMC6171242 DOI: 10.1186/s13690-018-0303-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/07/2018] [Indexed: 11/24/2022]
Abstract
Background Food insecurity is a global problem affecting many people worldwide, including approximately 220 million people in sub-Saharan Africa. Ethiopia is among the countries severely affected by hunger. However, evidence on how populations within Ethiopia cope with hunger and food insecurity is limited. This study aimed to identify household coping mechanisms in response to food insecurity at a Dabat Health and Demographic Surveillance System site. Methods This study used data from a re-census collected between October 2014 and December 2014.15,159 household members in thirteen kebeles of the Dabat Health and Demographic surveillance system were included. The outcome variables of the study were food insecurity and coping strategies. Household Food Insecurity Access Scale (HFIAS) was used to assess food insecurity. If food insecurity was found, families were asked about coping mechanisms used. Binary logistic regression analysis was applied to identify socio-demographic determinants of reducing amount and frequency of meal as a coping mechanism in response to food insecurity. Result Of the 15,159 households surveyed, 6671 (44.01%) reported the presence of a food insecurity in their household. Decreasing meal frequency and portions (3733 (55.96%)), borrowing money and food (2542 (38.11%)), and receiving food and money aid (1779 (26.67%)) were among the major coping strategies used by the households. Urban dwellers (AOR 2.07: 95% CI 1.74, 2.46), mid-altitude (weyina-dega) and high-land (dega) dwellers (AOR 2.46: 95% CI 2.08, 2.92 and AOR 1.22 95% CI 1.08, 1.38 respectively), and not married persons (AOR 1.60: 95% CI 1.07, 2.39) were more likely to consume less when faced with a food insecurity (using reducing amount and frequency of meal as a coping strategy). Conclusion Households in the study area experienced a very high rate of food insecurity. Decreasing meal frequency and portions was the primary coping mechanism used by the households. Due to the severe insecurity of food in their household, many people chose to reduce the amount and frequency of their meal in order to prolong the small amount of food in their house. This finding indicates a high risk for undernourishment which can exacerbate the burden of malnutrition and related diseases in the region.
Collapse
|
128
|
Human intestinal parasites in Mahajanga, Madagascar: The kingdom of the protozoa. PLoS One 2018; 13:e0204576. [PMID: 30304028 PMCID: PMC6179227 DOI: 10.1371/journal.pone.0204576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 09/11/2018] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Intestinal parasitic infections are a major public health problem in inter-tropical areas. The aim of our study was to describe the situation in Mahajanga, Madagascar with a particular focus on two protozoa, Dientamoeba fragilis and Blastocystis sp. METHODS This was a prospective study from February to June 2015. Stool samples from symptomatic hospitalized patients and asymptomatic volunteers were submitted to microscopy and molecular assays in order to detect parasites. RESULTS A wide panel of intestinal parasites were identified among the 265 included subjects, protozoa being the most prevalent with 72.8% whereas the prevalence of helminths and microsporidia was of 7.9% and 4.5%, respectively. Blastocystis sp. was the most prevalent protozoa (64.5% of the entire cohort) followed by various amoebas (35.5%) and flagellates (27,5%). We only detected subtypes 1, 2 and 3 of Blastocystis sp. Among the patients positive for D. fragilis (9.4%), 23 carried genotype 1 and 1 genotype 2. For the first time, we detected in 4 human stools the DNA of a recently described protozoon, Simplicimonas similis. Interestingly, subjects living in urban areas harbored significantly more different parasitic species than subjects living in rural areas with a correlation between sanitary level of neighborhood and protozoan infection. However, there was no difference in prevalence of digestive symptoms between parasite-free and parasite-infected subjects, except for Giardia intestinalis which had more symptomatic carriers. DISCUSSION Our study reveals a high overall parasite prevalence, similar to what had been found in 2003 in the same city and to other prevalence studies conducted in Africa. The poor access of the population to sanitary infrastructures may explain this result. Data from our study provide valuable key for sanitation programs and prevention of fecal-related infectious diseases.
Collapse
|
129
|
Platts-Mills JA, Liu J, Rogawski ET, Kabir F, Lertsethtakarn P, Siguas M, Khan SS, Praharaj I, Murei A, Nshama R, Mujaga B, Havt A, Maciel IA, McMurry TL, Operario DJ, Taniuchi M, Gratz J, Stroup SE, Roberts JH, Kalam A, Aziz F, Qureshi S, Islam MO, Sakpaisal P, Silapong S, Yori PP, Rajendiran R, Benny B, McGrath M, McCormick BJJ, Seidman JC, Lang D, Gottlieb M, Guerrant RL, Lima AAM, Leite JP, Samie A, Bessong PO, Page N, Bodhidatta L, Mason C, Shrestha S, Kiwelu I, Mduma ER, Iqbal NT, Bhutta ZA, Ahmed T, Haque R, Kang G, Kosek MN, Houpt ER. Use of quantitative molecular diagnostic methods to assess the aetiology, burden, and clinical characteristics of diarrhoea in children in low-resource settings: a reanalysis of the MAL-ED cohort study. LANCET GLOBAL HEALTH 2018; 6:e1309-e1318. [PMID: 30287127 PMCID: PMC6227251 DOI: 10.1016/s2214-109x(18)30349-8] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022]
Abstract
Background Optimum management of childhood diarrhoea in low-resource settings has been hampered by insufficient data on aetiology, burden, and associated clinical characteristics. We used quantitative diagnostic methods to reassess and refine estimates of diarrhoea aetiology from the Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) cohort study. Methods We re-analysed stool specimens from the multisite MAL-ED cohort study of children aged 0–2 years done at eight locations (Dhaka, Bangladesh; Vellore, India; Bhaktapur, Nepal; Naushero Feroze, Pakistan; Venda, South Africa; Haydom, Tanzania; Fortaleza, Brazil; and Loreto, Peru), which included active surveillance for diarrhoea and routine non-diarrhoeal stool collection. We used quantitative PCR to test for 29 enteropathogens, calculated population-level pathogen-specific attributable burdens, derived stringent quantitative cutoffs to identify aetiology for individual episodes, and created aetiology prediction scores using clinical characteristics. Findings We analysed 6625 diarrhoeal and 30 968 non-diarrhoeal surveillance stools from 1715 children. Overall, 64·9% of diarrhoea episodes (95% CI 62·6–71·2) could be attributed to an aetiology by quantitative PCR compared with 32·8% (30·8–38·7) using the original study microbiology. Viral diarrhoea (36·4% of overall incidence, 95% CI 33·6–39·5) was more common than bacterial (25·0%, 23·4–28·4) and parasitic diarrhoea (3·5%, 3·0–5·2). Ten pathogens accounted for 95·7% of attributable diarrhoea: Shigella (26·1 attributable episodes per 100 child-years, 95% CI 23·8–29·9), sapovirus (22·8, 18·9–27·5), rotavirus (20·7, 18·8–23·0), adenovirus 40/41 (19·0, 16·8–23·0), enterotoxigenic Escherichia coli (18·8, 16·5–23·8), norovirus (15·4, 13·5–20·1), astrovirus (15·0, 12·0–19·5), Campylobacter jejuni or C coli (12·1, 8·5–17·2), Cryptosporidium (5·8, 4·3–8·3), and typical enteropathogenic E coli (5·4, 2·8–9·3). 86·2% of the attributable incidence for Shigella was non-dysenteric. A prediction score for shigellosis was more accurate (sensitivity 50·4% [95% CI 46·7–54·1], specificity 84·0% [83·0–84·9]) than current guidelines, which recommend treatment only of bloody diarrhoea to cover Shigella (sensitivity 14·5% [95% CI 12·1–17·3], specificity 96·5% [96·0–97·0]). Interpretation Quantitative molecular diagnostics improved estimates of pathogen-specific burdens of childhood diarrhoea in the community setting. Viral causes predominated, including a substantial burden of sapovirus; however, Shigella had the highest overall burden with a high incidence in the second year of life. These data could improve the management of diarrhoea in these low-resource settings. Funding Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth T Rogawski
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - Shaila S Khan
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | | | | | - Buliga Mujaga
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | | | | | - Timothy L McMurry
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Darwin J Operario
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Jean Gratz
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Suzanne E Stroup
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - James H Roberts
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | | | | | | | - M Ohedul Islam
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Pimmada Sakpaisal
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Sasikorn Silapong
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Pablo P Yori
- Asociación Benéfica PRISMA, Iquitos, Peru; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | | | - Monica McGrath
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Jessica C Seidman
- Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | - Dennis Lang
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | - Michael Gottlieb
- Foundation for the National Institutes of Health, Bethesda, MD, USA
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | - Nicola Page
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Ladaporn Bodhidatta
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Carl Mason
- Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Sanjaya Shrestha
- Walter Reed/AFRIMS Research Unit, Nepal, Kathmandu, Nepal; University of Bergen, Bergen, Norway
| | - Ireen Kiwelu
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | | | | | | | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | - Margaret N Kosek
- Asociación Benéfica PRISMA, Iquitos, Peru; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
130
|
Khalil IA, Troeger C, Blacker BF, Rao PC, Brown A, Atherly DE, Brewer TG, Engmann CM, Houpt ER, Kang G, Kotloff KL, Levine MM, Luby SP, MacLennan CA, Pan WK, Pavlinac PB, Platts-Mills JA, Qadri F, Riddle MS, Ryan ET, Shoultz DA, Steele AD, Walson JL, Sanders JW, Mokdad AH, Murray CJL, Hay SI, Reiner RC. Morbidity and mortality due to shigella and enterotoxigenic Escherichia coli diarrhoea: the Global Burden of Disease Study 1990-2016. THE LANCET. INFECTIOUS DISEASES 2018; 18:1229-1240. [PMID: 30266330 PMCID: PMC6202441 DOI: 10.1016/s1473-3099(18)30475-4] [Citation(s) in RCA: 399] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023]
Abstract
Background Shigella and enterotoxigenic Escherichia coli (ETEC) are bacterial pathogens that are frequently associated with diarrhoeal disease, and are a significant cause of mortality and morbidity worldwide. The Global Burden of Diseases, Injuries, and Risk Factors study 2016 (GBD 2016) is a systematic, scientific effort to quantify the morbidity and mortality due to over 300 causes of death and disability. We aimed to analyse the global burden of shigella and ETEC diarrhoea according to age, sex, geography, and year from 1990 to 2016. Methods We modelled shigella and ETEC-related mortality using a Bayesian hierarchical modelling platform that evaluates a wide range of covariates and model types on the basis of vital registration and verbal autopsy data. We used a compartmental meta-regression tool to model the incidence of shigella and ETEC, which enforces an association between incidence, prevalence, and remission on the basis of scientific literature, population representative surveys, and health-care data. We calculated 95% uncertainty intervals (UIs) for the point estimates. Findings Shigella was the second leading cause of diarrhoeal mortality in 2016 among all ages, accounting for 212 438 deaths (95% UI 136 979–326 913) and about 13·2% (9·2–17·4) of all diarrhoea deaths. Shigella was responsible for 63 713 deaths (41 191–93 611) among children younger than 5 years and was frequently associated with diarrhoea across all adult age groups, increasing in elderly people, with broad geographical distribution. ETEC was the eighth leading cause of diarrhoea mortality in 2016 among all age groups, accounting for 51 186 deaths (26 757–83 064) and about 3·2% (1·8–4·7) of diarrhoea deaths. ETEC was responsible for about 4·2% (2·2–6·8) of diarrhoea deaths in children younger than 5 years. Interpretation The health burden of bacterial diarrhoeal pathogens is difficult to estimate. Despite existing prevention and treatment options, they remain a major cause of morbidity and mortality globally. Additional emphasis by public health officials is needed on a reduction in disease due to shigella and ETEC to reduce disease burden. Funding Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
| | | | | | - Puja C Rao
- Institute for Health Metrics and Evaluation, Seattle WA, USA
| | | | | | - Thomas G Brewer
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, USA
| | - Cyril M Engmann
- Maternal, Newborn, Child Health & Nutrition, PATH, Seattle, WA, USA; Department of Global Health, University of Washington School of Public Health, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Gagandeep Kang
- Translational Health Science and Technology Institute, Faridabad, India
| | - Karen L Kotloff
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Myron M Levine
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stephen P Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Calman A MacLennan
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - William K Pan
- Institute for Health Metrics and Evaluation, Seattle WA, USA; Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Patricia B Pavlinac
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Firdausi Qadri
- Infectious Diseases Division, International Center for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Edward T Ryan
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Department of Immunology & Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - David A Shoultz
- Drug Development, PATH, Seattle, WA, USA; Department of Global Health, University of Washington School of Public Health, Seattle, WA, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA; Albers School of Business & Economics, Seattle University, Seattle, WA, USA
| | - A Duncan Steele
- Enteric and Diarrheal Diseases, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Judd L Walson
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA; Department of Epidemiology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W Sanders
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ali H Mokdad
- Institute for Health Metrics and Evaluation, Seattle WA, USA
| | | | - Simon I Hay
- Institute for Health Metrics and Evaluation, Seattle WA, USA; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Robert C Reiner
- Institute for Health Metrics and Evaluation, Seattle WA, USA.
| |
Collapse
|
131
|
Carvalho MF, Gill D. Rotavirus vaccine efficacy: current status and areas for improvement. Hum Vaccin Immunother 2018; 15:1237-1250. [PMID: 30215578 PMCID: PMC6663136 DOI: 10.1080/21645515.2018.1520583] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/12/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
The difference noted in Rotavirus vaccine efficiency between high and low income countries correlates with the lack of universal access to clean water and higher standards of hygiene. Overcoming these obstacles will require great investment and also time, therefore more effective vaccines should be developed to meet the needs of those who would benefit the most from them. Increasing our current knowledge of mucosal immunity, response to Rotavirus infection and its modulation by circadian rhythms could point at actionable pathways to improve vaccination efficacy, especially in the case of individuals affected by environmental enteropathy. Also, a better understanding and validation of Rotavirus entry factors as well as the systematic monitoring of dominant strains could assist in tailoring vaccines to individual's needs. Another aspect that could improve vaccine efficiency is targeting to M cells, for which new ligands could potentially be sought. Finally, alternative mucosal adjuvants and vaccine expression, storage and delivery systems could have a positive impact in the outcome of Rotavirus vaccination.
Collapse
Affiliation(s)
| | - Davinder Gill
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| |
Collapse
|
132
|
Diarrhoeagenic Escherichia coli and Escherichia albertii in Brazil: pathotypes and serotypes over a 6-year period of surveillance. Epidemiol Infect 2018; 147:e10. [PMID: 30229714 PMCID: PMC6518528 DOI: 10.1017/s0950268818002595] [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] [Indexed: 02/07/2023] Open
Abstract
Diarrhoeagenic Escherichia coli (DEC) is a leading cause of infectious diarrhoea worldwide. In recent years, Escherichia albertii has also been implicated as a cause of human enteric diseases. This study describes the occurrence of E. coli pathotypes and serotypes associated with enteric illness and haemolytic uremic syndrome (HUS) isolated in Brazil from 2011 to 2016. Pathotypes isolated included enteropathogenic E. coli (EPEC), enteroaggregative E. coli (EAEC), enterotoxigenic E. coli (ETEC), enteroinvasive E. coli (EIEC) and Shiga toxin-producing E. coli (STEC). PCR of stool enrichments for DEC pathotypes was employed, and E. albertii was also sought. O:H serotyping was performed on all DEC isolates. A total of 683 DEC and 10 E. albertii strains were isolated from 5047 clinical samples. The frequencies of DEC pathotypes were 52.6% (359/683) for EPEC, 32.5% for EAEC, 6.3% for ETEC, 4.4% for EIEC and 4.2% for STEC. DEC strains occurred in patients from 3 months to 96 years old, but EPEC, EAEC and STEC were most prevalent among children. Both typical and atypical isolates of EPEC and EAEC were recovered and presented great serotype heterogeneity. HUS cases were only associated with STEC serotype O157:H7. Two E. albertii isolates belonged to serogroup O113 and one had the stx2f gene. The higher prevalence of atypical EPEC in relation to EAEC in community-acquired diarrhoea in Brazil suggests a shift in the trend of DEC pathotypes circulation as previously EAEC predominated. This is the first report of E. albertii isolation from active surveillance. These results highlight the need of continuing DEC and E. albertii surveillance, as a mean to detect changes in the pattern of pathotypes and serotypes circulation and provide useful information for intervention and control strategies.
Collapse
|
133
|
Chen C, Riley RT, Wu F. Dietary Fumonisin and Growth Impairment in Children and Animals: A Review. Compr Rev Food Sci Food Saf 2018; 17:1448-1464. [DOI: 10.1111/1541-4337.12392] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/24/2018] [Accepted: 08/01/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Chen Chen
- Dept. of Food Science and Human Nutrition; Michigan State Univ.; East Lansing MI 48824 U.S.A
- Inst. of Quality Standards and Testing Technology for Agro-Products; Chinese Academy of Agricultural Sciences; Beijing 100081 China
| | - Ronald T. Riley
- Dept. of Environmental Health Science; Univ. of Georgia; Athens GA 30602 U.S.A
| | - Felicia Wu
- Dept. of Food Science and Human Nutrition; Michigan State Univ.; East Lansing MI 48824 U.S.A
| |
Collapse
|
134
|
Biomarkers of Systemic Inflammation and Growth in Early Infancy are Associated with Stunting in Young Tanzanian Children. Nutrients 2018; 10:nu10091158. [PMID: 30149537 PMCID: PMC6164697 DOI: 10.3390/nu10091158] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/15/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
Stunting can afflict up to one-third of children in resource-constrained countries. We hypothesized that low-grade systemic inflammation (defined as elevations in serum C-reactive protein or alpha-1-acid glycoprotein) in infancy suppresses the growth hormone–insulin-like growth factor (IGF) axis and is associated with subsequent stunting. Blood samples of 590 children from periurban Dar es Salaam, Tanzania, were obtained at 6 weeks and 6 months of age as part of a randomized controlled trial. Primary outcomes were stunting, underweight, and wasting (defined as length-for-age, weight-for-age and weight-for-length z-scores < −2) between randomization and endline (18 months after randomization). Cox proportional hazards models were constructed to estimate hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) of time to first stunting, underweight, and wasting as outcomes, with measures of systemic inflammation, insulin-like growth factor-1 (IGF-1) and insulin-like growth factor binding protein-3 (IGFBP-3) as exposures, adjusting for numerous demographic and clinical variables. The incidences of subsequent stunting, underweight, and wasting were 26%, 20%, and 18%, respectively. In multivariate analyses, systemic inflammation at 6 weeks of age was significantly associated with stunting (HR: 2.14, 95% CI: 1.23, 3.72; p = 0.002). Children with higher levels of IGF-1 at 6 weeks were less likely to become stunted (HR: 0.58, 95% CI: 0.37, 0.93; p for trend = 0.019); a similar trend was noted in children with higher levels of IGF-1 at 6 months of age (HR: 0.50, 95% CI: 0.22, 1.12; p for trend = 0.07). Systemic inflammation occurs as early as 6 weeks of age and is associated with the risk of future stunting among Tanzanian children.
Collapse
|
135
|
Ives R, Humphrey L. Endochondral growth disruption during vitamin D deficiency rickets in a mid-19th century series from Bethnal Green, London, UK. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 167:585-601. [DOI: 10.1002/ajpa.23687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/23/2018] [Accepted: 07/02/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Rachel Ives
- Department of Earth Sciences; Natural History Museum; London The United Kingdom
- AOC Archaeology Group; Twickenham The United Kingdom
| | - Louise Humphrey
- Department of Earth Sciences; Natural History Museum; London The United Kingdom
| |
Collapse
|
136
|
Kinyoki DK, Moloney GM, Uthman OA, Odundo EO, Kandala NB, Noor AM, Snow RW, Berkley JA. Co-morbidity of malnutrition with falciparum malaria parasitaemia among children under the aged 6-59 months in Somalia: a geostatistical analysis. Infect Dis Poverty 2018; 7:72. [PMID: 29986753 PMCID: PMC6036667 DOI: 10.1186/s40249-018-0449-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 06/07/2018] [Indexed: 11/10/2022] Open
Abstract
Background Malnutrition and malaria are both significant causes of morbidity and mortality in African children. However, the extent of their spatial comorbidity remains unexplored and an understanding of their spatial correlation structure would inform improvement of integrated interventions. We aimed to determine the spatial correlation between both wasting and low mid upper arm circumference (MUAC) and falciparum malaria among Somalian children aged 6–59 months. Methods Data were from 49 227 children living in 888 villages between 2007 to 2010. We developed a Bayesian geostatistical shared component model in order to determine the common spatial distributions of wasting and falciparum malaria; and low-MUAC and falciparum malaria at 1 × 1 km spatial resolution. Results The empirical correlations with malaria were 0.16 and 0.23 for wasting and low-MUAC respectively. Shared spatial residual effects were statistically significant for both wasting and low-MUAC. The posterior spatial relative risk was highest for low-MUAC and malaria (range: 0.19 to 5.40) and relatively lower between wasting and malaria (range: 0.11 to 3.55). Hotspots for both wasting and low-MUAC with malaria occurred in the South Central region in Somalia. Conclusions The findings demonstrate a relationship between nutritional status and falciparum malaria parasitaemia, and support the use of the relatively simpler MUAC measurement in surveys. Shared spatial distribution and distinct hotspots present opportunities for targeted seasonal chemoprophylaxis and other forms of malaria prevention integrated within nutrition programmes. Electronic supplementary material The online version of this article (10.1186/s40249-018-0449-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Damaris K Kinyoki
- Spatial Health Metrics Group, INFORM Project, Kenya Medical Research Institute/Wellcome Trust Research Programme, Nairobi, Kenya.
| | - Grainne M Moloney
- Nutrition Section, United Nations Children's Fund (UNICEF), Kenya Country Office, UN Complex Gigiri, Nairobi, Kenya
| | - Olalekan A Uthman
- Warwick Medical School, Health Sciences Research Institute, Warwick Evidence, University of Warwick, Gibbet Hill, Coventry, CV4 7AL, UK
| | - Elijah O Odundo
- Food Security and Nutrition Analysis Unit (FSNAU) - Somalia, Food and Agriculture Organization of the United Nations, Ngecha Road Campus, Nairobi, Kenya
| | - Ngianga-Bakwin Kandala
- Department of Mathematics and Information sciences, Faculty of Engineering and Environment, Northumbria University, Newcastle upon Tyne, UK.,Faculty of Health and Sport Sciences, University of Agder, Kristiansand, Norway.,Division of Epidemiology and Biostatistics, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Abdisalan M Noor
- Spatial Health Metrics Group, INFORM Project, Kenya Medical Research Institute/Wellcome Trust Research Programme, Nairobi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, CCVTM, Oxford, OX3 7LJ, UK
| | - Robert W Snow
- Spatial Health Metrics Group, INFORM Project, Kenya Medical Research Institute/Wellcome Trust Research Programme, Nairobi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, CCVTM, Oxford, OX3 7LJ, UK
| | - James A Berkley
- Kenya Medical Research Institute/ Wellcome Trust Research Programme, Centre for Geographic Medicine Research (coast), Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, CCVTM, Oxford, OX3 7LJ, UK
| |
Collapse
|
137
|
Dalvi PS, Yang S, Swain N, Kim J, Saha S, Bourdon C, Zhang L, Chami R, Bandsma RHJ. Long-term metabolic effects of malnutrition: Liver steatosis and insulin resistance following early-life protein restriction. PLoS One 2018; 13:e0199916. [PMID: 29965973 PMCID: PMC6028108 DOI: 10.1371/journal.pone.0199916] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Early postnatal-life malnutrition remains prevalent globally, and about 45% of all child deaths are linked to malnutrition. It is not clear whether survivors of childhood malnutrition suffer from long-term metabolic effects, especially when they are later in life exposed to a fat and carbohydrate rich obesogenic diet. The lack of knowledge around this dietary “double burden” warrants studies to understand the long-term consequences of children previously exposed to malnutrition. We hypothesized that an early-life nutritional insult of low protein consumption in mice would lead to long-term metabolic disturbances that would exacerbate the development of diet-induced insulin resistance and non-alcoholic fatty liver disease (NAFLD). We investigated the effects of feeding a low protein diet (4% wt/wt) immediately after weaning for four weeks and subsequent feeding of a high carbohydrate high fat feeding for 16 weeks on metabolic function and development of NAFLD. Mice exposed to early-life protein restriction demonstrated a transient glucose intolerance upon recovery by regular chow diet feeding. However, protein restriction after weaning in mice did not exacerbate an obesogenic diet-induced insulin resistance or progression to NAFLD. These data suggest that transient protein restriction in early-life does not exacerbate an obesogenic diet-induced NAFLD and insulin resistance.
Collapse
Affiliation(s)
- Prasad S. Dalvi
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Morosky College of Health Professions and Sciences, Gannon University, Erie, PA, United States of America
| | - Steven Yang
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nathan Swain
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Junsoo Kim
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Senjuti Saha
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Celine Bourdon
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ling Zhang
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rose Chami
- Department of Laboratory Medicine and Pathology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Robert H. J. Bandsma
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
138
|
Pit Latrines: A Noninvasive Sampling Strategy to Assess Fecal Pathogen Occurrence in Low Resource Communities. J Community Health 2018; 43:1155-1160. [PMID: 29948524 DOI: 10.1007/s10900-018-0535-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Limited understanding of disease in low resource communities continues to hamper improvements in health. We evaluated household pit latrine sampling as a non-invasive approach to investigate important fecal pathogens such as Giardia lamblia and Cryptosporidium spp. in impoverished communities where health-seeking behavior limits the sensitivity of health facility-based surveillance. Fecal samples were collected from pit latrines in randomly selected households and from patients presenting to the only hospital in the region during the same time periods. Samples were tested with a commercially available ELISA. Giardia household prevalence was 28.7% in 2016 and 48.4% in 2017, while individual samples from hospital submission had a Giardia prevalence of 2.4% in 2016 and 8.0% in 2017. Cryptosporidium was only found in one household. Results suggest that pit latrine surveillance for fecal-borne infections provide course estimates of community infection levels that are unbiased by health seeking behaviors and allow surveillance of vulnerable sectors of a population.
Collapse
|
139
|
Kumar A, Vlasova AN, Deblais L, Huang HC, Wijeratne A, Kandasamy S, Fischer DD, Langel SN, Paim FC, Alhamo MA, Shao L, Saif LJ, Rajashekara G. Impact of nutrition and rotavirus infection on the infant gut microbiota in a humanized pig model. BMC Gastroenterol 2018; 18:93. [PMID: 29929472 PMCID: PMC6013989 DOI: 10.1186/s12876-018-0810-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 05/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human rotavirus (HRV) is a major cause of viral gastroenteritis in infants; particularly in developing countries where malnutrition is prevalent. Malnutrition perturbs the infant gut microbiota leading to sub-optimal functioning of the immune system and further predisposing infants to enteric infections. Therefore, we hypothesized that malnutrition exacerbates rotavirus disease severity in infants. METHODS In the present study, we used a neonatal germ free (GF) piglets transplanted with a two-month-old human infant's fecal microbiota (HIFM) on protein deficient and sufficient diets. We report the effects of malnourishment on the HRV infection and the HIFM pig microbiota in feces, intestinal and systemic tissues, using MiSeq 16S gene sequencing (V4-V5 region). RESULTS Microbiota analysis indicated that the HIFM transplantation resulted in a microbial composition in pigs similar to that of the original infant feces. This model was then used to understand the interconnections between microbiota diversity, diet, and HRV infection. Post HRV infection, HIFM pigs on the deficient diet had lower body weights, developed more severe diarrhea and increased virus shedding compared to HIFM pigs on sufficient diet. However, HRV induced diarrhea and shedding was more pronounced in non-colonized GF pigs compared to HIFM pigs on either sufficient or deficient diet, suggesting that the microbiota alone moderated HRV infection. HRV infected pigs on sufficient diet showed increased microbiota diversity in intestinal tissues; whereas, greater diversity was observed in systemic tissues of HRV infected pigs fed with deficient diet. CONCLUSIONS These results suggest that proper nourishment improves the microbiota quality in the intestines, alleviates HRV disease and lower probability of systemic translocation of potential opportunistic pathogens/pathobionts. In conclusion, our findings further support the role for microbiota and proper nutrition in limiting enteric diseases.
Collapse
Affiliation(s)
- Anand Kumar
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Group B-10: Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM USA
| | - Anastasia N. Vlasova
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Loic Deblais
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Huang-Chi Huang
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Asela Wijeratne
- The Molecular and Cellular Imaging Center, The Ohio State University, Wooster, OH USA
| | - Sukumar Kandasamy
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - David D. Fischer
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Stephanie N. Langel
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Francine Chimelo Paim
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Moyasar A. Alhamo
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Lulu Shao
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Hillman Cancer Center, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260 USA
| | - Linda J. Saif
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Gireesh Rajashekara
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| |
Collapse
|
140
|
Critical Role of Zinc in a New Murine Model of Enterotoxigenic Escherichia coli Diarrhea. Infect Immun 2018; 86:IAI.00183-18. [PMID: 29661930 PMCID: PMC6013668 DOI: 10.1128/iai.00183-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of traveler's diarrhea as well as of endemic diarrhea and stunting in children in developing areas. However, a small-mammal model has been badly needed to better understand and assess mechanisms, vaccines, and interventions. We report a murine model of ETEC diarrhea, weight loss, and enteropathy and investigate the role of zinc in the outcomes. ETEC strains producing heat-labile toxins (LT) and heat-stable toxins (ST) that were given to weaned C57BL/6 mice after antibiotic disruption of normal microbiota caused growth impairment, watery diarrhea, heavy stool shedding, and mild to moderate intestinal inflammation, the latter being worse with zinc deficiency. Zinc treatment promoted growth in zinc-deficient infected mice, and subinhibitory levels of zinc reduced expression of ETEC virulence genes cfa1, cexE, sta2, and degP but not of eltA in vitro Zinc supplementation increased shedding and the ileal burden of wild-type (WT) ETEC but decreased shedding and the tissue burden of LT knockout (LTKO) ETEC. LTKO ETEC-infected mice had delayed disease onset and also had less inflammation by fecal myeloperoxidase (MPO) assessment. These findings provide a new murine model of ETEC infection that can help elucidate mechanisms of growth, diarrhea, and inflammatory responses as well as potential vaccines and interventions.
Collapse
|
141
|
Troeger C, Colombara DV, Rao PC, Khalil IA, Brown A, Brewer TG, Guerrant RL, Houpt ER, Kotloff KL, Misra K, Petri WA, Platts-Mills J, Riddle MS, Swartz SJ, Forouzanfar MH, Reiner RC, Hay SI, Mokdad AH. Global disability-adjusted life-year estimates of long-term health burden and undernutrition attributable to diarrhoeal diseases in children younger than 5 years. LANCET GLOBAL HEALTH 2018; 6:e255-e269. [PMID: 29433665 PMCID: PMC5861379 DOI: 10.1016/s2214-109x(18)30045-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 12/12/2022]
Abstract
Background Diarrhoea is a leading cause of death and illness globally among children younger than 5 years. Mortality and short-term morbidity cause substantial burden of disease but probably underestimate the true effect of diarrhoea on population health. This underestimation is because diarrhoeal diseases can negatively affect early childhood growth, probably through enteric dysfunction and impaired uptake of macronutrients and micronutrients. We attempt to quantify the long-term sequelae associated with childhood growth impairment due to diarrhoea. Methods We used the Global Burden of Diseases, Injuries, and Risk Factors Study framework and leveraged existing estimates of diarrhoea incidence, childhood undernutrition, and infectious disease burden to estimate the effect of diarrhoeal diseases on physical growth, including weight and height, and subsequent disease among children younger than 5 years. The burden of diarrhoea was measured in disability-adjusted life-years (DALYs), a composite metric of mortality and morbidity. We hypothesised that diarrhoea is negatively associated with three common markers of growth: weight-for-age, weight-for-height, and height-for-age Z-scores. On the basis of these undernutrition exposures, we applied a counterfactual approach to quantify the relative risk of infectious disease (subsequent diarrhoea, lower respiratory infection, and measles) and protein energy malnutrition morbidity and mortality per day of diarrhoea and quantified the burden of diarrhoeal disease due to these outcomes caused by undernutrition. Findings Diarrhoea episodes are significantly associated with childhood growth faltering. We found that each day of diarrhoea was associated with height-for-age Z-score (–0·0033 [95% CI −0·0024 to −0·0041]; p=4·43 × 10−14), weight-for-age Z-score (–0·0077 [–0·0058 to −0·0097]; p=3·19 × 10−15), and weight-for-height Z-score (–0·0096 [–0·0067 to −0·0125]; p=7·78 × 10−11). After addition of the DALYs due to the long-term sequelae as a consequence of undernutrition, the burden of diarrhoeal diseases increased by 39·0% (95% uncertainty interval [UI] 33·0–46·6) and was responsible for 55 778 000 DALYs (95% UI 49 125 400–62 396 200) among children younger than 5 years in 2016. Among the 15 652 300 DALYs (95% UI 12 951 300–18 806 100) associated with undernutrition due to diarrhoeal episodes, more than 84·7% are due to increased risk of infectious disease, whereas the remaining 15·3% of long-term DALYs are due to increased prevalence of protein energy malnutrition. The burden of diarrhoea has decreased substantially since 1990, but progress has been greater in long-term (78·7% reduction [95% UI 69·3–85·5]) than in acute (70·4% reduction [95% UI 61·7–76·5]) DALYs. Interpretation Diarrhoea represents an even larger burden of disease than was estimated in the Global Burden of Disease Study. In order to adequately address the burden of its long-term sequelae, a renewed emphasis on controlling the risk of diarrhoea incidence may be required. This renewed effort can help further prevent the potential lifelong cost on child health, growth, and overall potential. Funding Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Christopher Troeger
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Danny V Colombara
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Puja C Rao
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Ibrahim A Khalil
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Alexandria Brown
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | | | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Karen L Kotloff
- Departments of Pediatrics and Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kavita Misra
- Bureau of HIV/AIDS Prevention and Control, New York City Department of Health and Mental Hygiene, New York, NY, USA
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - James Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Mark S Riddle
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Scott J Swartz
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | | | - Robert C Reiner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Simon I Hay
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Ali H Mokdad
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA.
| |
Collapse
|
142
|
Chen C, Mitchell NJ, Gratz J, Houpt ER, Gong Y, Egner PA, Groopman JD, Riley RT, Showker JL, Svensen E, Mduma ER, Patil CL, Wu F. Exposure to aflatoxin and fumonisin in children at risk for growth impairment in rural Tanzania. ENVIRONMENT INTERNATIONAL 2018; 115:29-37. [PMID: 29544138 PMCID: PMC5989662 DOI: 10.1016/j.envint.2018.03.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/01/2018] [Accepted: 03/01/2018] [Indexed: 05/11/2023]
Abstract
Growth impairment is a major public health issue for children in Tanzania. The question remains as to whether dietary mycotoxins play a role in compromising children's growth. We examined children's exposures to dietary aflatoxin and fumonisin and potential impacts on growth in 114 children under 36 months of age in Haydom, Tanzania. Plasma samples collected from the children at 24 months of age (N = 60) were analyzed for aflatoxin B1-lysine (AFB1-lys) adducts, and urine samples collected between 24 and 36 months of age (N = 94) were analyzed for urinary fumonisin B1 (UFB1). Anthropometric, socioeconomic, and nutritional parameters were measured and growth parameter z-scores were calculated for each child. Seventy-two percent of the children had detectable levels of AFB1-lys, with a mean level of 5.1 (95% CI: 3.5, 6.6) pg/mg albumin; and 80% had detectable levels of UFB1, with a mean of 1.3 (95% CI: 0.8, 1.8) ng/ml. This cohort had a 75% stunting rate [height-for-age z-scores (HAZ) < -2] for children at 36 months. No associations were found between aflatoxin exposures and growth impairment as measured by stunting, underweight [weight-for-age z-scores (WAZ) < -2], or wasting [weight-for-height z-scores (WHZ) < -2]. However, fumonisin exposure was negatively associated with underweight (with non-detectable samples included, p = 0.0285; non-detectable samples excluded, p = 0.005) in this cohort of children. Relatively low aflatoxin exposure at 24 months was not linked with growth impairment, while fumonisin exposure at 24-36 months based on the UFB1 biomarkers may contribute to the high growth impairment rate among children of Haydom, Tanzania; which may be associated with their breast feeding and weaning practices.
Collapse
Affiliation(s)
- Chen Chen
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Nicole J Mitchell
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA; Uckele Health and Nutrition, Blissfield, MI, USA
| | - Jean Gratz
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Yunyun Gong
- School of Food Science and Nutrition, University of Leeds, UK
| | - Patricia A Egner
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - John D Groopman
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ronald T Riley
- Toxicology and Mycotoxin Research Unit, National Poultry Disease Research Center, R.B. Russell Research Center, USDA-ARS, Athens, GA, USA
| | - Jency L Showker
- Toxicology and Mycotoxin Research Unit, National Poultry Disease Research Center, R.B. Russell Research Center, USDA-ARS, Athens, GA, USA
| | - Erling Svensen
- Haukeland University Hospital, Bergen, Norway; Haydom Lutheran Hospital, Manyara Region, Tanzania; University of Bergen, Norway
| | | | - Crystal L Patil
- Department of Women, Children and Family Health Science, University of Illinois at Chicago, College of Nursing, Chicago, IL, USA
| | - Felicia Wu
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
143
|
Markers of Environmental Enteric Dysfunction Are Associated With Neurodevelopmental Outcomes in Tanzanian Children. J Pediatr Gastroenterol Nutr 2018; 66:953-959. [PMID: 29613921 PMCID: PMC5964017 DOI: 10.1097/mpg.0000000000001978] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chronic exposure to enteropathogens may result in environmental enteric dysfunction (EED), a subclinical condition associated with poor child growth. Growth faltering is strongly associated with poor neurodevelopment, and occurs during sensitive periods of postnatal brain development. We investigated the role of novel EED biomarkers, systemic inflammation, and micronutrient status on neurodevelopment in Tanzanian children. METHODS Non-stunted subjects with 6-week and 6-month blood samples and neurodevelopmental measures (n = 107) were included in this study. Samples were tested for biomarkers of gastrointestinal function (citrulline, antibodies to lipopolysaccharide, and flagellin), micronutrient status (iron, retinol binding protein [RBP], and vitamin D), systemic inflammation (C-reactive protein [CRP] and alpha-1-acid glycoprotein), and growth (insulin-like growth factor and insulin-like growth factor binding protein 3). RESULTS Cognitive scores at 15 months were associated with higher concentrations of 6-month anti-lipopolysaccharide IgG (β = 1.95, P = 0.02), anti-flagellin IgA (β = 2.41, P = 0.04), and IgG (β = 2.99, P = 0.009). Higher receptive language scores were positively associated with anti-flagellin IgG (β = 0.95, P = 0.05), and receptive language and gross motor scores were positively associated with citrulline at 6 months (β = 0.09, P = 0.02; β = 0.10, P = 0.03, respectively). Gross motor scores were positively associated with RBP at 6 months (β = 1.70, P = 0.03). Markers of systemic inflammation were not significantly associated with neurodevelopment. CONCLUSIONS Plasma citrulline, a marker of gastrointestinal mucosal surface area, and vitamin A status were associated with higher gross motor development scores. Novel markers for EED, but not inflammation, were positively associated with cognitive scores, suggesting a possible mechanistic pathway involving immune response and neuroprotection.
Collapse
|
144
|
Gaur S, Lopez EC, Ojha A, Andrade JE. Functionalization of Lipid-Based Nutrient Supplement with β-Cyclodextrin Inclusions of Oregano Essential Oil. J Food Sci 2018; 83:1748-1756. [PMID: 29771453 DOI: 10.1111/1750-3841.14178] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 04/01/2018] [Accepted: 04/06/2018] [Indexed: 12/17/2022]
Abstract
Intestinal parasitic infection is one of the main causes of acute undernutrition in children. Oral consumption of oregano essential oil (OEO) can reduce intestinal parasitic infections, however, its addition to therapeutic and supplementary foods is hampered by its undesirable flavor. The objective of this study was to develop a functional lipid-based nutrient supplement (LNS) containing OEO, which is stable, acceptable and provides targeted intestinal delivery of bioactive. β-cyclodextrin (β-CyD) inclusion complexes of OEO (β-CyD-OEO), and carvacrol (β-CyD-CV) (1:1 molar) were prepared using slurry complexation (-20 °C) method and characterized based on encapsulation efficiency, moisture content, morphology, and 2-phase in vitro digestion stability. Carvacrol (CV) content was measured using reverse phase HPLC-UV. LNS containing β-CyD-OEO (27.2 mg encapsulate/20 g LNS) was formulated using Indian staples and ingredients. Discriminatory sensory tests (triangle) were performed with college students (n = 58) and low-income women (n = 25), with young children at home (1 to 6 years), living in Mehsana, India to evaluate differences between LNS with and without bioactive ingredient (β-CyD-OEO only). Moisture of dried complexes ranged 9.1% to 9.7% d.b., whereas water activity 0.35 to 0.412. The complex size and encapsulation efficiency of β-CyD-OEO and β-CyD-CV were 1.5 to 7 μm and 4 to 20 μm, and 86.04 ± 4.48% and 81.39 ± 3.34%, respectively. The bioactive complexes were stable through the gastric and intestinal phases. Bioaccessibility of encapsulated CV ranged 6.0% to 7.7%. Sensory tests revealed no differences (P > 0.05) in color, aroma, and taste between LNS with and without β-CyD-OEO complexes. Functionalization of LNS with β-CyD-OEO is feasible based on in vitro stability and sensory studies. PRACTICAL APPLICATION Despite its antiparasitic activities, the addition of oregano essential oil into foods is limited due to its strong flavor and volatility. In this study, we evaluated the encapsulation of oregano essential oil with β-cyclodextrin and its addition into lipid-based nutrition supplements. The results revealed that complex encapsulation efficiency was above 80%. Also, the bioactive complexes were stable under in vitro gastrointestinal conditions. Sensory evaluation of LNS with and without encapsulated essential oil showed no difference in terms of color, aroma, and taste. The functional LNS can both address nutrient insufficiency as well as parasitic infection among malnourished populations in low-resource settings.
Collapse
Affiliation(s)
- Shashank Gaur
- Dept. of Food Science and Human Nutrition, Univ. of Illinois at Urbana-Champaign, Urbana, IL 61801, U.S.A
| | - Emely C Lopez
- Dept. of Food Science and Human Nutrition, Univ. of Illinois at Urbana-Champaign, Urbana, IL 61801, U.S.A
| | - Ankur Ojha
- Dept. of Food Science and Technology, Natl. Inst. of Food Technology Entrepreneurship and Management, Sonipat, Haryana 131028, India
| | - Juan E Andrade
- Dept. of Food Science and Human Nutrition, Univ. of Illinois at Urbana-Champaign, Urbana, IL 61801, U.S.A.,Div. of Nutritional Sciences, Univ. of Illinois at Urbana-Champaign, Urbana, IL 61801, U.S.A
| |
Collapse
|
145
|
Ramteke SM, Shiau S, Foca M, Strehlau R, Pinillos F, Patel F, Violari A, Liberty A, Coovadia A, Kuhn L, Arpadi SM. Patterns of Growth, Body Composition, and Lipid Profiles in a South African Cohort of Human Immunodeficiency Virus-Infected and Uninfected Children: A Cross-Sectional Study. J Pediatric Infect Dis Soc 2018; 7:143-150. [PMID: 28481997 PMCID: PMC5954242 DOI: 10.1093/jpids/pix026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/14/2017] [Indexed: 11/14/2022]
Abstract
BACKGROUND Prior research in sub-Saharan Africa reports dyslipidemia in perinatally human immunodeficiency virus (HIV)-infected children receiving ritonavir-boosted lopinavir (LPV/r) compared with efavirenz; however, interpretation of findings is limited by lack of comparison data from HIV-uninfected children. METHODS We conducted a cross-sectional analysis of lipid profiles and growth within a larger longitudinal cohort study of perinatally HIV-infected and HIV-uninfected children aged 4-9 years in Johannesburg, South Africa. At enrollment, anthropometrics, viral load, CD4, total cholesterol (TC), high-density lipoprotein, low-density lipoprotein (LDL), and triglycerides were measured. Weight-for-age Z-score (WAZ), height-for-age Z-score (HAZ), and body mass index-for-age Z-score (BAZ) were calculated. United States pediatric thresholds for dyslipidemia were used. RESULTS Five hundred fifty-three HIV-infected and 300 HIV-uninfected children (median age 6.9 years) of similar demographic characteristics were enrolled. Of the HIV-infected children, 94.8% were on combination antiretroviral therapy (cART) (65.4% on LPV/r- and 28.6% on efavirenz-based regimens). Among the treated, 94.3% had a viral load <200 copies/mL. Median CD4% was 34.4. The HIV-infected children had lower mean WAZ (-0.7 vs -0.3, P < .01) and HAZ (-1.1 vs -0.7, P < .01) compared with HIV-uninfected children. A lower proportion of HIV-infected children were overweight (BAZ >1) compared with HIV-uninfected children (14.4% vs 21.7%, P = .04). Whether on LPV/r or efavirenz, a higher proportion of HIV-infected children had borderline/elevated TC or abnormal triglycerides than HIV-uninfected children, although a higher proportion of those on LPV/r had borderline/elevated TC, borderline/elevated LDL, or abnormal triglycerides than those on efavirenz. CONCLUSIONS In a South African cohort of HIV-infected children and population-appropriate HIV-uninfected children, unfavorable alterations in lipid profiles were detected in HIV-infected children regardless of treatment regimen compared with HIV-uninfected children. The HIV-infected children were of smaller size than HIV-uninfected children, but there was a high prevalence of overweight in both groups. Strategies for optimizing growth and early life management of lipid alterations may be warranted.
Collapse
Affiliation(s)
- Sarah M Ramteke
- Gertrude H. Sergievsky Center, Columbia University, New York
| | - Stephanie Shiau
- Gertrude H. Sergievsky Center, Columbia University, New York,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York,Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marc Foca
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Francoise Pinillos
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Faeezah Patel
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Avy Violari
- Perinatal HIV Research Unit, Chris Hani Baragwanath Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Afaaf Liberty
- Perinatal HIV Research Unit, Chris Hani Baragwanath Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Ashraf Coovadia
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Center, Columbia University, New York,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York,Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephen M Arpadi
- Gertrude H. Sergievsky Center, Columbia University, New York,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York,Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York,Perinatal HIV Research Unit, Chris Hani Baragwanath Hospital, University of the Witwatersrand, Johannesburg, South Africa,Correspondence: S. M. Arpadi, MD, MS, Gertrude H. Sergievsky Center, Columbia University, 622 W. 168th Street, PH 19—114, New York, NY 10032 ()
| | | |
Collapse
|
146
|
Garzón M, Pereira-da-Silva L, Seixas J, Papoila AL, Alves M. Subclinical Enteric Parasitic Infections and Growth Faltering in Infants in São Tomé, Africa: A Birth Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E688. [PMID: 29621166 PMCID: PMC5923730 DOI: 10.3390/ijerph15040688] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023]
Abstract
The associations between enteric pathogenic parasites and growth in infants in São Tomé were explored using a refined anthropometric approach to recognize early growth faltering. A birth cohort study was conducted with follow-up to 24 months of age. Microscopic examination for protozoa and soil-transmitted helminths was performed. Anthropometric assessments included: z-scores for weight-for-length (WLZ), length-for-age (LAZ), weight (WAVZ) and length velocities (LAVZ), length-for-age difference (LAD), and wasting and stunting risk (≤-1 SD). Generalized additive mixed effects regression models were used to explore the associations between anthropometric parameters and enteric parasitic infections and cofactors. A total of 475 infants were enrolled, and 282 completed the study. The great majority of infants were asymptomatic. Giardia lamblia was detected in 35.1% of infants in at least one stool sample, helminths in 30.4%, and Cryptosporidium spp. in 14.7%. Giardia lamblia and helminth infections were significantly associated with mean decreases of 0.10 in LAZ and 0.32 in LAD, and of 0.16 in LAZ and 0.48 in LAD, respectively. Cryptosporidium spp. infection was significantly associated with a mean decrease of 0.43 in WAVZ and 0.55 in LAVZ. The underestimated association between subclinical parasitic enteric infections and mild growth faltering in infants should be addressed in public health policies.
Collapse
Affiliation(s)
- Marisol Garzón
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa; 1349-008 Lisbon, Portugal.
- Global Health and Tropical Medicine R&D Center, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa; 1349-008 Lisbon, Portugal.
| | - Luís Pereira-da-Silva
- Medicine of Woman, Childhood and Adolescence Teaching and Research Area, NOVA Medical School, Universidade NOVA de Lisboa; 1169-056 Lisbon, Portugal.
- Research Unit, Centro Hospitalar de Lisboa Central; 1169-045 Lisbon, Portugal.
| | - Jorge Seixas
- Tropical Clinic Teaching and Research Unit, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa; 1349-008 Lisbon, Portugal.
- Global Health and Tropical Medicine R&D Center, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa; 1349-008 Lisbon, Portugal.
| | - Ana Luísa Papoila
- Research Unit, Centro Hospitalar de Lisboa Central; 1169-045 Lisbon, Portugal.
| | - Marta Alves
- Research Unit, Centro Hospitalar de Lisboa Central; 1169-045 Lisbon, Portugal.
| |
Collapse
|
147
|
Environmental enteric dysfunction and systemic inflammation predict reduced weight but not length gain in rural Bangladeshi children. Br J Nutr 2018; 119:407-414. [DOI: 10.1017/s0007114517003683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractEnvironmental enteric dysfunction (EED) and systemic inflammation (SI) are common in developing countries and may cause stunting. In Bangladesh, >40 % of preschool children are stunted, but EED and SI contributions are unknown. We aimed to determine the impact of EED and SI (assessed with multiple indicators) on growth in children (n 539) enrolled in a community-based randomised food supplementation trial in rural Bangladesh. EED was defined with faecal myeloperoxidase, α-1 antitrypsin and neopterin and serum endotoxin core antibody and glucagon-like peptide-2, consolidated into gut inflammation (GI) and permeability (GP) scores, and urinary lactulose:mannitol α-1 acid glycoprotein (AGP) characterised SI. Biomarker associations with anthropometry (15-, 18- and 24-month length-for-age (LAZ), weight-for-length (WLZ) and weight-for-age (WAZ) z scores) were examined in pairwise correlations and adjusted mixed-effects regressions. Stunting, wasting and underweight prevalence at 18 months were 45, 15 and 37 %, respectively, with elevated EED and SI markers common. EED and SI were not associated with 15–24-month length trajectory. Elevated (worse) GI and GP scores predicted reduced 18–24-month WLZ change (β −0·01 (se 0·00) z score/month for both). Elevated GP was also associated with reduced 15–18-month WLZ change (β −0·03 (se 0·01) z score/month) and greater 15-month WLZ (β 0·16 (se 0·05)). Higher AGP was associated with reduced prior and increased subsequent WLZ change (β −0·04 (se 0·01) and β 0·02 (se 0·00) z score/month for 15–18 and 18–24 months). The hypothesised link from EED to stunting was not observed in this sample of Bangladeshi 18-month-olds, but the effects of EED on constrained weight gain may have consequences for later linear growth or for other health and development outcomes.
Collapse
|
148
|
Scharf RJ, Rogawski ET, Murray-Kolb LE, Maphula A, Svensen E, Tofail F, Rasheed M, Abreu C, Vasquez AO, Shrestha R, Pendergast L, Mduma E, Koshy B, Conaway MR, Platts-Mills JA, Guerrant RL, DeBoer MD. Early childhood growth and cognitive outcomes: Findings from the MAL-ED study. MATERNAL AND CHILD NUTRITION 2018; 14:e12584. [PMID: 29392824 DOI: 10.1111/mcn.12584] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/10/2017] [Accepted: 11/27/2017] [Indexed: 01/08/2023]
Abstract
Although many studies around the world hope to measure or improve developmental progress in children to promote community flourishing and productivity, growth is sometimes used as a surrogate because cognitive skills are more difficult to measure. Our objective was to assess how childhood measures of anthropometry correlate with measures of child development in low-income settings with high prevalence of poor nutrition and enteric disease, to inform studies considering growth outcomes in the absence of direct child developmental skill assessment. Children from the MAL-ED study were followed from birth to 24 months of age in field sites in 8 low- and middle-income countries across 3 continents. Monthly weight, length, and head circumference measurements were performed. At 24 months, the Bayley Scales of Infant and Toddler Development was administered. We correlated cognitive measures at 24 months with anthropometric measurements from birth to 2 years comparing 3 constructs: absolute attained monthly measures, summative difference in measures from the mean growth curve, and rate of change in measures. Growth faltering at multiple time periods is related to Bayley cognitive outcomes at 24 months. Birthweight, overall growth by 18-24 months, and rate of growth in the 6- to 18-month period were most associated with 24-month developmental scores. In this study, head circumference measurements, compared with length, was more closely linked to cognitive scores at 24 months. Notably, all studies between growth and cognitive outcomes exhibited low r2 values (0.001-0.049). Anthropometric measures, particularly head circumference, were related to cognitive development, although explaining a low percent of variance. When feasible, direct measures of child development may be more useful.
Collapse
Affiliation(s)
- Rebecca J Scharf
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA.,Center for Global Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth T Rogawski
- Center for Global Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Laura E Murray-Kolb
- Department of Nutrition Sciences, Penn State University, University Park, Pennsylvania, USA
| | - Angelina Maphula
- Department of Psychology, University of Venda, Thohoyandou, South Africa
| | - Erling Svensen
- Department of Global Health and Primary Care, University of Bergen, Bergen, Norway
| | - Fahmida Tofail
- Center for Nutrition and Food Security, icddr-b, Dhaka, Bangladesh
| | - Muneera Rasheed
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Claudia Abreu
- Department of Microbiology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Rita Shrestha
- Department of Psychology, Siddhi Memorial Hospital, Bhaktapur, Nepal
| | - Laura Pendergast
- Department of Psychology, Temple University, Philadelphia, Pennsylvania, USA
| | - Estomih Mduma
- Haydom Global Health Research Centre, Haydom, Tanzania
| | - Beena Koshy
- Department of Developmental Pediatrics, Christian Medical College, Vellore, India
| | - Mark R Conaway
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - James A Platts-Mills
- Center for Global Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Richard L Guerrant
- Center for Global Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Mark D DeBoer
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
149
|
Wagner VE, Dey N, Guruge J, Hsiao A, Ahern PP, Semenkovich NP, Blanton LV, Cheng J, Griffin N, Stappenbeck TS, Ilkayeva O, Newgard CB, Petri W, Haque R, Ahmed T, Gordon JI. Effects of a gut pathobiont in a gnotobiotic mouse model of childhood undernutrition. Sci Transl Med 2017; 8:366ra164. [PMID: 27881825 DOI: 10.1126/scitranslmed.aah4669] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022]
Abstract
To model how interactions among enteropathogens and gut microbial community members contribute to undernutrition, we colonized gnotobiotic mice fed representative Bangladeshi diets with sequenced bacterial strains cultured from the fecal microbiota of two 24-month-old Bangladeshi children: one healthy and the other underweight. The undernourished donor's bacterial collection contained an enterotoxigenic Bacteroides fragilis strain (ETBF), whereas the healthy donor's bacterial collection contained two nontoxigenic strains of B. fragilis (NTBF). Analyses of mice harboring either the unmanipulated culture collections or systematically manipulated versions revealed that ETBF was causally related to weight loss in the context of its native community but not when introduced into the healthy donor's community. This phenotype was transmissible from the dams to their offspring and was associated with derangements in host energy metabolism manifested by impaired tricarboxylic acid cycle activity and decreased acyl-coenzyme A utilization. NTBF reduced ETBF's expression of its enterotoxin and mitigated the effects of ETBF on the transcriptomes of other healthy donor community members. These results illustrate how intraspecific (ETBF-NTBF) and interspecific interactions influence the effects of harboring B. fragilis.
Collapse
Affiliation(s)
- Vitas E Wagner
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Neelendu Dey
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janaki Guruge
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ansel Hsiao
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip P Ahern
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas P Semenkovich
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura V Blanton
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiye Cheng
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas Griffin
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710, USA.,Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - William Petri
- Departments of Medicine, Microbiology, and Pathology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka 1212, Bangladesh
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka 1212, Bangladesh
| | - Jeffrey I Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA. .,Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
150
|
Relationship between growth and illness, enteropathogens and dietary intakes in the first 2 years of life: findings from the MAL-ED birth cohort study. BMJ Glob Health 2017; 2:e000370. [PMID: 29333282 PMCID: PMC5759708 DOI: 10.1136/bmjgh-2017-000370] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/04/2017] [Accepted: 09/17/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Dietary and illness factors affect risk of growth faltering; the role of enteropathogens is less clear. As part of the Etiology, Risk Factors and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) study, we quantify the effects of enteropathogen infection, diarrhoea and diet on child growth. METHODS Newborns were enrolled and followed until 24 months. Length and weight were assessed monthly. Illnesses and breastfeeding practices were documented biweekly; from 9 to 24 months, non-breast milk intakes were quantified monthly. Routinely collected non-diarrhoeal stools were analysed for a broad array of enteropathogens. A linear piecewise spline model was used to quantify associations of each factor with growth velocity in seven of eight MAL-ED sites; cumulative effects on attained size at 24 months were estimated for mean, low (10th percentile) and high (90th percentile) exposure levels. Additionally, the six most prevalent enteropathogens were evaluated for their effects on growth. RESULTS Diarrhoea did not have a statistically significant effect on growth. Children with high enteropathogen exposure were estimated to be 1.21±0.33 cm (p<0.001; 0.39 length for age (LAZ)) shorter and 0.08±0.15 kg (p=0.60; 0.08 weight-for-age (WAZ)) lighter at 24 months, on average, than children with low exposure. Campylobacter and enteroaggregativeEscherichia coli detections were associated with deficits of 0.83±0.33 and 0.85±0.31 cm in length (p=0.011 and 0.001) and 0.22±0.15 and 0.09±0.14 kg in weight (p=0.14 and 0.52), respectively. Children with low energy intakes and protein density were estimated to be 1.39±0.33 cm (p<0.001; 0.42 LAZ) shorter and 0.81±0.15 kg (p<0.001; 0.65 WAZ) lighter at 24 months than those with high intakes. CONCLUSIONS Reducing enteropathogen burden and improving energy and protein density of complementary foods could reduce stunting.
Collapse
|