51
|
Pronovost GN, Hsiao EY. Perinatal Interactions between the Microbiome, Immunity, and Neurodevelopment. Immunity 2019; 50:18-36. [PMID: 30650376 PMCID: PMC6447295 DOI: 10.1016/j.immuni.2018.11.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/17/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
The microbiome modulates host immune function across the gastrointestinal tract, peripheral lymphoid organs, and central nervous system. In this review, we highlight emerging evidence that microbial effects on select immune phenotypes arise developmentally, where the maternal and neonatal microbiome influence immune cell ontogeny in the offspring during gestation and early postnatal life. We further discuss roles for the perinatal microbiome and early-life immunity in regulating normal neurodevelopmental processes. In addition, we examine evidence that abnormalities in microbiota-neuroimmune interactions during early life are associated with altered risk of neurological disorders in humans. Finally, we conclude by evaluating the potential implications of microbiota-immune interventions for neurological conditions. Continued progress toward dissecting mechanistic interactions between the perinatal microbiota, immune system, and nervous system might uncover fundamental insights into how developmental interactions across physiological systems inform later-life health and disease.
Collapse
Affiliation(s)
- Geoffrey N Pronovost
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
52
|
|
53
|
Cox LA, Olivier M, Spradling-Reeves K, Karere GM, Comuzzie AG, VandeBerg JL. Nonhuman Primates and Translational Research-Cardiovascular Disease. ILAR J 2018; 58:235-250. [PMID: 28985395 DOI: 10.1093/ilar/ilx025] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the United States. Human epidemiological studies provide challenges for understanding mechanisms that regulate initiation and progression of CVD due to variation in lifestyle, diet, and other environmental factors. Studies describing metabolic and physiologic aspects of CVD, and those investigating genetic and epigenetic mechanisms influencing CVD initiation and progression, have been conducted in multiple Old World nonhuman primate (NHP) species. Major advantages of NHPs as models for understanding CVD are their genetic, metabolic, and physiologic similarities with humans, and the ability to control diet, environment, and breeding. These NHP species are also genetically and phenotypically heterogeneous, providing opportunities to study gene by environment interactions that are not feasible in inbred animal models. Each Old World NHP species included in this review brings unique strengths as models to better understand human CVD. All develop CVD without genetic manipulation providing multiple models to discover genetic variants that influence CVD risk. In addition, as each of these NHP species age, their age-related comorbidities such as dyslipidemia and diabetes are accelerated proportionally 3 to 4 times faster than in humans.In this review, we discuss current CVD-related research in NHPs focusing on selected aspects of CVD for which nonprimate model organism studies have left gaps in our understanding of human disease. We include studies on current knowledge of genetics, epigenetics, calorie restriction, maternal calorie restriction and offspring health, maternal obesity and offspring health, nonalcoholic steatohepatitis and steatosis, Chagas disease, microbiome, stem cells, and prevention of CVD.
Collapse
Affiliation(s)
- Laura A Cox
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Michael Olivier
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | | | - Genesio M Karere
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - John L VandeBerg
- South Texas Diabetes and Obesity Center, School of Medicine, University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, Texas
| |
Collapse
|
54
|
Miklavcic JJ, Badger TM, Bowlin AK, Matazel KS, Cleves MA, LeRoith T, Saraf MK, Chintapalli SV, Piccolo BD, Shankar K, Yeruva L. Human Breast-Milk Feeding Enhances the Humoral and Cell-Mediated Immune Response in Neonatal Piglets. J Nutr 2018; 148:1860-1870. [PMID: 30247686 PMCID: PMC6209812 DOI: 10.1093/jn/nxy170] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 06/22/2018] [Accepted: 07/03/2018] [Indexed: 12/24/2022] Open
Abstract
Background The benefits of breastfeeding infants are well characterized, including those on the immune system. However, determining the mechanism by which human breast milk (HBM) elicits effects on immune response requires investigation in an appropriate animal model. Objective The primary aim of this study was to develop a novel porcine model and to determine the differential effects of feeding HBM and a commercial milk formula (MF) on immune response and gastrointestinal microbial colonization in a controlled environment. Methods Male piglets were fed HBM (n = 26) or MF (n = 26) from day 2 through day 21. Piglets were vaccinated (n = 9/diet group) with cholera toxin and cholera toxin subunit B (CTB) and tetanus toxoid at 21 d or were fed placebo (n = 6/diet group) and then weaned to a standard solid diet at the age of 21 d. Humoral and cell-mediated immune responses were assessed from blood on days 35 and 48. Immune response was further examined from tissues, including mesenteric lymph nodes (MLNs), Peyer's patches (PPs), and spleen. The colonization of gut microbiota was characterized from feces on days 16 and 49. Results Serum antibody titers in piglets fed HBM were 4-fold higher (P < 0.05) to CTB and 3-fold higher (P < 0.05) to tetanus toxoid compared with piglets fed MF on day 48. Compared with MF, the numbers of immunoglobulin A antibody-producing cells to CTB were 13-fold higher (P < 0.05) in MLNs and 11-fold higher (P < 0.05) in PPs in the HBM diet group on day 51. In addition, significantly increased T cell proliferation was observed in the HBM group relative to the MF group. Furthermore, microbial diversity in the HBM group was lower (P < 0.05) than in the MF group. Conclusions This porcine model appears to be valid for studying the effects of early postnatal diet on immune responses and the gastrointestinal microbiome. Our results lay the groundwork for future studies defining the role of infant diet on microbiota and immune function.
Collapse
Affiliation(s)
- John J Miklavcic
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Katelin S Matazel
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Mario A Cleves
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
- Arkansas Children's Research Institute, Little Rock, AR
| | - Tanya LeRoith
- Department of Biomedical Science and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Manish K Saraf
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
- Arkansas Children's Research Institute, Little Rock, AR
| |
Collapse
|
55
|
Abstract
The human body is colonized by a diverse collective of microorganisms, including bacteria, fungi, protozoa and viruses. The smallest entity of this microbial conglomerate are the bacterial viruses. Bacteriophages, or phages for short, exert significant selective pressure on their bacterial hosts, undoubtedly influencing the human microbiome and its impact on our health and well-being. Phages colonize all niches of the body, including the skin, oral cavity, lungs, gut, and urinary tract. As such our bodies are frequently and continuously exposed to diverse collections of phages. Despite the prevalence of phages throughout our bodies, the extent of their interactions with human cells, organs, and immune system is still largely unknown. Phages physically interact with our mucosal surfaces, are capable of bypassing epithelial cell layers, disseminate throughout the body and may manipulate our immune system. Here, I establish the novel concept of an "intra-body phageome," which encompasses the collection of phages residing within the classically "sterile" regions of the body. This review will take a phage-centric view of the microbiota, human body, and immune system with the ultimate goal of inspiring a greater appreciation for both the indirect and direct interactions between bacteriophages and their mammalian hosts.
Collapse
Affiliation(s)
- Jeremy J Barr
- School of Biological Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
56
|
Laing ST, Merriam D, Shock BC, Mills S, Spinner A, Reader R, Hartigan-O'Connor DJ. Idiopathic Colitis in Rhesus Macaques Is Associated With Dysbiosis, Abundant Enterochromaffin Cells and Altered T-Cell Cytokine Expression. Vet Pathol 2018; 55:741-752. [PMID: 29929446 DOI: 10.1177/0300985818780449] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Idiopathic chronic diarrhea (ICD) is a common ailment affecting captive rhesus macaques ( Macaca mulatta). ICD cases are characterized by diarrhea in the absence of commonly identified diarrheal pathogens and multiple recurrences even after supportive therapy. Histologically, the disease is characterized by lymphoplasmacytic colitis. We identified 35 rhesus macaques euthanized for ICD during a 7-month period and described demographic, clinical, histologic, and immunologic commonalities. We found a trend of historic Campylobacter spp. and trichomonad infections. Furthermore, rhesus macaques with ICD demonstrated loss of normal colonic adherent bacterium, identified in this study as Helicobacter macacae; increased abundance of Pentatrichomonas hominis; and increased frequency of colonic serotonin-positive enterochromaffin cells. Interestingly, colonic and ileal T-helper cells of animals with ICD manifested decreased capacity for expression of certain cytokines, in particular interleukin (IL)-4 and IL-13. These data further describe a common ailment and suggest new avenues to identify complex interactions involved in the etiology of recurring diarrhea in young rhesus macaques.
Collapse
Affiliation(s)
| | - David Merriam
- 2 California National Primate Research Center, University of California, Davis, CA, USA.,3 Department of Medical Microbiology and Immunology, University of California, Davis, CA, USA
| | - Barbara C Shock
- 4 Department of Biology, Lincoln Memorial University, Harrogate, TN, USA
| | - Sarah Mills
- 2 California National Primate Research Center, University of California, Davis, CA, USA
| | - Abbie Spinner
- 2 California National Primate Research Center, University of California, Davis, CA, USA
| | - Rachel Reader
- 2 California National Primate Research Center, University of California, Davis, CA, USA
| | - Dennis J Hartigan-O'Connor
- 2 California National Primate Research Center, University of California, Davis, CA, USA.,3 Department of Medical Microbiology and Immunology, University of California, Davis, CA, USA.,5 Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
57
|
Clayton JB, Gomez A, Amato K, Knights D, Travis DA, Blekhman R, Knight R, Leigh S, Stumpf R, Wolf T, Glander KE, Cabana F, Johnson TJ. The gut microbiome of nonhuman primates: Lessons in ecology and evolution. Am J Primatol 2018; 80:e22867. [PMID: 29862519 DOI: 10.1002/ajp.22867] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 03/23/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023]
Abstract
The mammalian gastrointestinal (GI) tract is home to trillions of bacteria that play a substantial role in host metabolism and immunity. While progress has been made in understanding the role that microbial communities play in human health and disease, much less attention has been given to host-associated microbiomes in nonhuman primates (NHPs). Here we review past and current research exploring the gut microbiome of NHPs. First, we summarize methods for characterization of the NHP gut microbiome. Then we discuss variation in gut microbiome composition and function across different NHP taxa. Finally, we highlight how studying the gut microbiome offers new insights into primate nutrition, physiology, and immune system function, as well as enhances our understanding of primate ecology and evolution. Microbiome approaches are useful tools for studying relevant issues in primate ecology. Further study of the gut microbiome of NHPs will offer new insight into primate ecology and evolution as well as human health.
Collapse
Affiliation(s)
- Jonathan B Clayton
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota.,GreenViet Biodiversity Conservation Center, Son Tra District, Danang, Vietnam.,Primate Microbiome Project, Minneapolis, Minnesota
| | - Andres Gomez
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Animal Science, University of Minnesota, St Paul, Minnesota
| | - Katherine Amato
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Anthropology, Northwestern University, Evanston, Illinois
| | - Dan Knights
- Primate Microbiome Project, Minneapolis, Minnesota.,Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota.,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Dominic A Travis
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Veterinary Population Medicine, University of Minnesota, Saint Paul, Minnesota
| | - Ran Blekhman
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota.,Department of Ecology, Evolution, and Behavior, University of Minnesota, Falcon Heights, Minnesota
| | - Rob Knight
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Computer Science & Engineering, UC San Diego, La Jolla, California.,Department of Pediatrics, UC San Diego, La Jolla, California.,Center for Microbiome Innovation, UC San Diego, La Jolla, California
| | - Steven Leigh
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Anthropology, University of Colorado Boulder, Boulder, Colorado.,C.R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois
| | - Rebecca Stumpf
- Primate Microbiome Project, Minneapolis, Minnesota.,C.R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois.,Department of Anthropology, University of Illinois, Urbana, Illinois
| | - Tiffany Wolf
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Veterinary Population Medicine, University of Minnesota, Saint Paul, Minnesota
| | - Kenneth E Glander
- Primate Microbiome Project, Minneapolis, Minnesota.,Department of Evolutionary Anthropology, Duke University, Durham, North Carolina
| | - Francis Cabana
- Primate Microbiome Project, Minneapolis, Minnesota.,Wildlife Nutrition Centre, Wildlife Reserves Singapore, Singapore
| | - Timothy J Johnson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota.,Primate Microbiome Project, Minneapolis, Minnesota.,University of Minnesota, Mid-Central Research and Outreach Center, Willmar, Minnesota
| |
Collapse
|
58
|
Nagpal R, Shively CA, Appt SA, Register TC, Michalson KT, Vitolins MZ, Yadav H. Gut Microbiome Composition in Non-human Primates Consuming a Western or Mediterranean Diet. Front Nutr 2018; 5:28. [PMID: 29922651 PMCID: PMC5996930 DOI: 10.3389/fnut.2018.00028] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/06/2018] [Indexed: 01/18/2023] Open
Abstract
The mammalian gastrointestinal tract harbors a highly diverse and dynamic community of bacteria. The array of this gut bacterial community, which functions collectively as a fully unified organ in the host metabolism, varies greatly among different host species and can be shaped by long-term nutritional interventions. Non-human primates, our close phylogenetic relatives and ancestors, provide an excellent model for studying diet-microbiome interaction; however, compared to clinical and rodent studies, research targeting primate gut microbiome has been limited. Herein, we analyze the gut microbiome composition in female cynomolgus macaques (Macaca fascicularis; n = 20) after the long-term (2.5 years) consumption of diets designed to mimic recent human Western- (WD; n = 10) or Mediterranean-type (MD; n = 10) diets. Microbiome diversity in MD consumers was significantly higher by the Shannon diversity index compared to the WD consumers, with similar but non-significant trends noted for the diversity metrics of species richness (Chao 1), observed operational taxonomic units (OTUs) and phylogenetic diversity (PD) whole Tree. Compared to the MD, the WD group demonstrated a higher Firmicutes-Bacteroides ratio and a significantly higher abundance of families Clostridiacea and Lactobacillaceae. Further analyses reveal significantly higher abundance of genera Lactobacillus, Clostridium, Faecalibacterium, and Oscillospira and lower abundance of Ruminococcus and Coprococcus in MD consumers relative to WD consumers. OTUs belonging to several species also show significant differences between the two groups, with Lactobacillus species demonstrating a prominently higher abundance in the MD consumers. The data reveal several differences in the gut microbiome of primates consuming the two different diets and should be useful for further studies aimed at understanding the diet-microbiome-health interactions in primates.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine and Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Carol A Shively
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Susan A Appt
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Thomas C Register
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Kristofer T Michalson
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mara Z Vitolins
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine and Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
59
|
Erturk-Hasdemir D, Kasper DL. Finding a needle in a haystack: Bacteroides fragilis polysaccharide A as the archetypical symbiosis factor. Ann N Y Acad Sci 2018. [PMID: 29528123 DOI: 10.1111/nyas.13660] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Starting from birth, all animals develop a symbiotic relationship with their resident microorganisms that benefits both the microbe and the host. Recent advances in technology have substantially improved our ability to direct research toward the identification of important microbial species that affect host physiology. The identification of specific commensal molecules from these microbes and their mechanisms of action is still in its early stages. Polysaccharide A (PSA) of Bacteroides fragilis is the archetypical example of a commensal molecule that can modulate the host immune system in health and disease. This zwitterionic polysaccharide has a critical impact on the development of the mammalian immune system and also on the stimulation of interleukin 10-producing CD4+ T cells; consequently, PSA confers benefits to the host with regard to experimental autoimmune, inflammatory, and infectious diseases. In this review, we summarize the current understanding of the immunomodulatory effects of B. fragilis PSA and discuss these effects as a novel immunological paradigm. In particular, we discuss recent advances in our understanding of the unique functional mechanisms of this molecule and its therapeutic potential, and we review the recent literature in the field of microbiome research aimed at discovering new commensal products and their immunomodulatory potential.
Collapse
Affiliation(s)
- Deniz Erturk-Hasdemir
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Dennis L Kasper
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
60
|
Le Doare K, Holder B, Bassett A, Pannaraj PS. Mother's Milk: A Purposeful Contribution to the Development of the Infant Microbiota and Immunity. Front Immunol 2018; 9:361. [PMID: 29599768 PMCID: PMC5863526 DOI: 10.3389/fimmu.2018.00361] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Breast milk is the perfect nutrition for infants, a result of millions of years of evolution. In addition to providing a source of nutrition, breast milk contains a diverse array of microbiota and myriad biologically active components that are thought to guide the infant’s developing mucosal immune system. It is believed that bacteria from the mother’s intestine may translocate to breast milk and dynamically transfer to the infant. Such interplay between mother and her infant is a key to establishing a healthy infant intestinal microbiome. These intestinal bacteria protect against many respiratory and diarrheal illnesses, but are subject to environmental stresses such as antibiotic use. Orchestrating the development of the microbiota are the human milk oligosaccharides (HMOs), the synthesis of which are partially determined by the maternal genotype. HMOs are thought to play a role in preventing pathogenic bacterial adhesion though multiple mechanisms, while also providing nutrition for the microbiome. Extracellular vesicles (EVs), including exosomes, carry a diverse cargo, including mRNA, miRNA, and cytosolic and membrane-bound proteins, and are readily detectable in human breast milk. Strongly implicated in cell–cell signaling, EVs could therefore may play a further role in the development of the infant microbiome. This review considers the emerging role of breast milk microbiota, bioactive HMOs, and EVs in the establishment of the neonatal microbiome and the consequent potential for modulation of neonatal immune system development.
Collapse
Affiliation(s)
- Kirsty Le Doare
- Centre for International Child Health, Imperial College London, London, United Kingdom.,Paediatrics, Imperial College London, London, United Kingdom.,Paediatric Infectious Diseases Research Group, St. George's, University of London, London, United Kingdom.,Vaccines & Immunity Theme, MRC Unit The Gambia, Fajara, Gambia
| | - Beth Holder
- Centre for International Child Health, Imperial College London, London, United Kingdom.,Paediatrics, Imperial College London, London, United Kingdom
| | - Aisha Bassett
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Pia S Pannaraj
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States.,Department of Pediatrics and Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
61
|
Moossavi S, Miliku K, Sepehri S, Khafipour E, Azad MB. The Prebiotic and Probiotic Properties of Human Milk: Implications for Infant Immune Development and Pediatric Asthma. Front Pediatr 2018; 6:197. [PMID: 30140664 PMCID: PMC6095009 DOI: 10.3389/fped.2018.00197] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022] Open
Abstract
The incidence of pediatric asthma has increased substantially in recent decades, reaching a worldwide prevalence of 14%. This rapid increase may be attributed to the loss of "Old Friend" microbes from the human microbiota resulting in a less diverse and "dysbiotic" gut microbiota, which fails to optimally stimulate immune development during infancy. This hypothesis is supported by observations that the gut microbiota is different in infants who develop asthma later in life compared to those who remain healthy. Thus, early life exposures that influence gut microbiota play a crucial role in asthma development. Breastfeeding is one such exposure; it is generally considered protective against pediatric asthma, although conflicting results have been reported, potentially due to variations in milk composition between individuals and across populations. Human milk oligosaccharides (HMOs) and milk microbiota are two major milk components that influence the infant gut microbiota and hence, development of the immune system. Among their many immunomodulatory functions, HMOs exert a selective pressure within the infant gut microbial niche, preferentially promoting the proliferation of specific bacteria including Bifidobacteria. Milk is also a source of viable bacteria originating from the maternal gut and infant oral cavity. As such, breastmilk has prebiotic and probiotic properties that can modulate two of the main forces controlling the gut microbial community assembly, i.e., dispersal and selection. Here, we review the latest evidence, mechanisms and hypotheses for the synergistic and/or additive effects of milk microbiota and HMOs in protecting against pediatric asthma.
Collapse
Affiliation(s)
- Shirin Moossavi
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kozeta Miliku
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Shadi Sepehri
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Ehsan Khafipour
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Meghan B Azad
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.,Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
62
|
Ximenez C, Torres J. Development of Microbiota in Infants and its Role in Maturation of Gut Mucosa and Immune System. Arch Med Res 2017; 48:666-680. [PMID: 29198451 DOI: 10.1016/j.arcmed.2017.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/15/2017] [Indexed: 12/24/2022]
Abstract
Dysbiosis of the gut microbiota has been associated with increasing numbers of diseases, including obesity, diabetes, inflammatory bowel disease, asthma, allergy, cancer and even neurologic or behavioral disorders. The other side of the coin is that a healthy microbiota leads to a healthy human development, to a mature and well trained immune system and to an efficient metabolic machinery. What we have learned in adults is in the end the result of a good start, a programmed, healthy development of the microbiota that must occur in the early years of life, probably even starting during the fetal stage. This review aims to present and discuss reports that helps us understand what we have learned of the development of microbiota during the early times of life, from pregnancy to delivery to the early years after birth. The impact of the establishment of "healthy" bacterial communities on human surfaces in the maturation of epithelia, immune system and metabolism will also be discussed. The right process of maturation of the bacterial communities that establish a symbiosis with human surfaces depends on a number of environmental, genetic and temporal factors that need to be understand in order to have tools to monitor a healthy development and eventually intervene to correct undesired courses.
Collapse
Affiliation(s)
- Cecilia Ximenez
- Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas, Unidad Médica de Alta Especialidad, Hospital de Pediatría, Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Ciudad de México, México.
| |
Collapse
|
63
|
Méndez-Lagares G, Lu D, Merriam D, Baker CA, Villinger F, Van Rompay KKA, McCune JM, Hartigan-O'Connor DJ. IL-21 Therapy Controls Immune Activation and Maintains Antiviral CD8 + T Cell Responses in Acute Simian Immunodeficiency Virus Infection. AIDS Res Hum Retroviruses 2017; 33:S81-S92. [PMID: 29140110 DOI: 10.1089/aid.2017.0160] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) replicate during acute infection in lymphocytes of the gastrointestinal tract, before disseminating systemically. Localized replication and associated loss of gut-resident CD4+ T cells occur regardless of the portal of entry of the virus (e.g., intravenous vs. rectal). Thus, HIV and SIV are tropic for gut tissue, and their pathogenesis requires the special environment of the intestine. T helper 17 (Th17) cells are important contributors to microbial defense in the gut that are vulnerable to HIV infection and whose loss is associated with translocation of microbial products to the systemic circulation, leading to chronic immune activation and disease progression. Interleukin (IL)-21 promotes differentiation and survival of Th17 cells and stimulates CD8+ T cell function. By promoting Th17 cell survival, IL-21 could limit bacterial translocation and immune activation in the setting of acute or rebounding HIV/SIV disease. In this study, we tested the effect of recombinant IL-21-IgFc treatment, given at the time of infection, on SIVmac251 infection. We found that rIL-21-IgFc decreases immune activation and maintains effective antiviral responses by CD8+ T cells in blood, but this maintenance is not associated with lower viral loads. rIL-21-IgFc treatment also did not generally support Th17 cell populations, but Th17 cells remained strongly and independently associated with control of plasma viremia. For example, the single animal exhibiting greatest control over viremia in our study also manifested the highest levels of IL-21 in plasma, Th17 cell maintenance in blood, and Th17 cells in intestinal tissue. These findings provide rationale for further exploration of IL-21 treatment as a support for host CD8+ T cell responses in HIV cure strategies.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - Ding Lu
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - David Merriam
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
| | - Christopher A. Baker
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - François Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California, Davis, California
| | - Joseph M. McCune
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| | - Dennis J. Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, California
- Department of Medical Microbiology and Immunology, University of California, Davis, California
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California
| |
Collapse
|
64
|
HIV DNA-Adenovirus Multiclade Envelope Vaccine Induces gp41 Antibody Immunodominance in Rhesus Macaques. J Virol 2017; 91:JVI.00923-17. [PMID: 28794027 DOI: 10.1128/jvi.00923-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/31/2017] [Indexed: 11/20/2022] Open
Abstract
Dominant antibody responses in vaccinees who received the HIV-1 multiclade (A, B, and C) envelope (Env) DNA/recombinant adenovirus virus type 5 (rAd5) vaccine studied in HIV-1 Vaccine Trials Network (HVTN) efficacy trial 505 (HVTN 505) targeted Env gp41 and cross-reacted with microbial antigens. In this study, we asked if the DNA/rAd5 vaccine induced a similar antibody response in rhesus macaques (RMs), which are commonly used as an animal model for human HIV-1 infections and for testing candidate HIV-1 vaccines. We also asked if gp41 immunodominance could be avoided by immunization of neonatal RMs during the early stages of microbial colonization. We found that the DNA/rAd5 vaccine elicited a higher frequency of gp41-reactive memory B cells than gp120-memory B cells in adult and neonatal RMs. Analysis of the vaccine-induced Env-reactive B cell repertoire revealed that the majority of HIV-1 Env-reactive antibodies in both adult and neonatal RMs were targeted to gp41. Interestingly, a subset of gp41-reactive antibodies isolated from RMs cross-reacted with host antigens, including autologous intestinal microbiota. Thus, gp41-containing DNA/rAd5 vaccine induced dominant gp41-microbiota cross-reactive antibodies derived from blood memory B cells in RMs as observed in the HVTN 505 vaccine efficacy trial. These data demonstrated that RMs can be used to investigate gp41 immunodominance in candidate HIV-1 vaccines. Moreover, colonization of neonatal RMs occurred within the first week of life, and immunization of neonatal RMs during this time also induced a dominant gp41-reactive antibody response.IMPORTANCE Our results are critical to current work in the HIV-1 vaccine field evaluating the phenomenon of gp41 immunodominance induced by HIV-1 Env gp140 in RMs and humans. Our data demonstrate that RMs are an appropriate animal model to study this phenomenon and to determine the immunogenicity in new HIV-1 Env trimer vaccine designs. The demonstration of gp41 immunodominance in memory B cells of both adult and neonatal RMs indicated that early vaccination could not overcome gp41 dominant responses.
Collapse
|
65
|
Amaral WZ, Lubach GR, Proctor A, Lyte M, Phillips GJ, Coe CL. Social Influences on Prevotella and the Gut Microbiome of Young Monkeys. Psychosom Med 2017; 79:888-897. [PMID: 28178033 PMCID: PMC5547018 DOI: 10.1097/psy.0000000000000454] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Our aim was to evaluate the bacterial profiles of young monkeys as they were weaned into peer groups with a particular focus on Prevotella, an important taxon in both human and nonhuman primates. The weaning of infants and increased social contact with peers is a developmental stage that is likely to affect the gut microbiome. METHODS Gut bacteria were assessed in 63 rhesus monkeys living in social groups comprised of 4 to 7 individuals. Two groups were assessed prospectively on day 1 and 2 weeks after rehousing away from the mother and group formation. Ten additional groups were assessed at 2 weeks after group establishment. Fecal genomic DNA was extracted and 16S ribosomal RNA sequenced by Illumina MiSeq (5 social groups) and 454-amplicon pyrosequencing (7 social groups). RESULTS Combining weaned infants into small social groups led to a microbial convergence by 2 weeks (p < .001). Diversity analyses indicated more similar community structure within peer groups than across groups (p < .01). Prevotella was the predominant taxon, and its abundance differed markedly across individuals. Indices of richness, microbial profiles, and less abundant taxa were all associated with the Prevotella levels. Functional Kyoto Encyclopedia of Genes and Genomes analyses suggested corresponding shifts in metabolic pathways. CONCLUSIONS The formation of small groups of young rhesus monkeys was associated with significant shifts in the gut microbiota. The profiles were closely associated with the abundance of Prevotella, a predominant taxon in the rhesus monkey gut. Changes in the structure of the gut microbiome are likely to induce differences in metabolic and physiologic functioning.
Collapse
Affiliation(s)
- Wellington Z Amaral
- From the Harlow Center for Biological Psychology (Amaral, Lubach, Coe), University of Wisconsin, Madison, Wisconsin; and the Department of Veterinary Microbiology and Veterinary Medicine (Proctor, Lyte, Phillips), Iowa State University, Ames, Iowa
| | | | | | | | | | | |
Collapse
|
66
|
Protecting the Newborn and Young Infant from Infectious Diseases: Lessons from Immune Ontogeny. Immunity 2017; 46:350-363. [PMID: 28329702 DOI: 10.1016/j.immuni.2017.03.009] [Citation(s) in RCA: 298] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/20/2016] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
Infections in the first year of life are common and often severe. The newborn host demonstrates both quantitative and qualitative differences to the adult in nearly all aspects of immunity, which at least partially explain the increased susceptibility to infection. Here we discuss how differences in susceptibility to infection result not out of a state of immaturity, but rather reflect adaptation to the particular demands placed on the immune system in early life. We review the mechanisms underlying host defense in the very young, and discuss how specific developmental demands increase the risk of particular infectious diseases. In this context, we discuss how this plasticity, i.e. the capacity to adapt to demands encountered in early life, also provides the potential to leverage protection of the young against infection and disease through a number of interventions.
Collapse
|
67
|
The microbiota in pediatric rheumatic disease: epiphenomenon or therapeutic target? Curr Opin Rheumatol 2017; 28:537-43. [PMID: 27286235 DOI: 10.1097/bor.0000000000000312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW There has been increasing interest in the contents and function of the microbiota, as it relates to pediatric inflammatory diseases. Here, we discuss the factors underlying the development of the microbiota, its role in juvenile idiopathic arthritis (JIA) and prospects for therapeutic interventions in the microbiota. RECENT FINDINGS The human microbiota undergoes a succession of changes, until it reaches a mature form. A variety of early-life exposures, including mode of delivery and form of feeding, can affect the contents of the microbiota and possibly impact upon long-term risk of developing autoimmune diseases. The microbiota is altered in children with JIA, including elevated Bacteroides genus in JIA as a whole and decreased Faecalibacterium prausnitzii in pediatric spondyloarthritis. Although there are limited data so far indicating that microbiota-based therapies can result in therapeutic improvement of arthritis, most of the data are on adults and thus may not be applicable to children. SUMMARY Perturbations of the microbiota during childhood may result in the development of a microbiota associated with increased risk of pediatric rheumatic illness. Whether the microbiota can be targeted is a focus of ongoing research.
Collapse
|
68
|
Saraf MK, Piccolo BD, Bowlin AK, Mercer KE, LeRoith T, Chintapalli SV, Shankar K, Badger TM, Yeruva L. Formula diet driven microbiota shifts tryptophan metabolism from serotonin to tryptamine in neonatal porcine colon. MICROBIOME 2017; 5:77. [PMID: 28705171 PMCID: PMC5513086 DOI: 10.1186/s40168-017-0297-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 07/04/2017] [Indexed: 05/06/2023]
Abstract
BACKGROUND The gut microbiota of breast-fed and formula-fed infants differ significantly, as do the risks for allergies, gut dysfunction, and upper respiratory tract infections. The connections between breast milk, various formulas, and the profiles of gut bacteria to these childhood illnesses, as well as the mechanisms underlying the effects, are not well understood. METHODS We investigated distal colon microbiota by 16S RNA amplicon sequencing, morphology by histomorphometry, immune response by cytokine expression, and tryptophan metabolism in a pig model in which piglets were sow-fed, or fed soy or dairy milk-based formula from postnatal day (PND) 2 to 21. RESULTS Formula feeding significantly (p < 0.05) altered the colon microbiota relative to the sow feeding. A significant reduction in microbial diversity was noted with formula groups in comparison to sow-fed. Streptococcus, Blautia, Citrobacter, Butrycimonas, Parabacteroides, Lactococcus genera were increased with formula feeding relative to sow feeding. In addition, relative to sow feeding, Anaerotruncus, Akkermansia, Enterococcus, Acinetobacter, Christensenella, and Holdemania were increased in milk-fed piglets, and Biliophila, Ruminococcus, Clostridium were increased in soy-fed piglets. No significant gut morphological changes were noted. However, higher cytokine mRNA expression (BMP4, CCL11, CCL21) was observed in the distal colon of formula groups. Formula feeding reduced enterochromaffin cell number and serotonin, but increased tryptamine levels relative to sow feeding. CONCLUSION Our data confirm that formula diet alters the colon microbiota and appears to shift tryptophan metabolism from serotonin to tryptamine, which may lead to greater histamine levels and risk of allergies in infants.
Collapse
Affiliation(s)
- Manish Kumar Saraf
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Brian D. Piccolo
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Anne K. Bowlin
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Kelly E. Mercer
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, USA
| | - Sree V. Chintapalli
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Kartik Shankar
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Thomas M. Badger
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Laxmi Yeruva
- Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
- Arkansas Children’s Research Institute, Little Rock, USA
| |
Collapse
|
69
|
Sidener HM, Park B, Gao L. Effect of Antibiotic Administration during Infancy on Growth Curves through Young Adulthood in Rhesus Macaques ( Macaca mulatta). Comp Med 2017; 67:270-276. [PMID: 28662756 PMCID: PMC5482519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/23/2016] [Accepted: 12/06/2016] [Indexed: 06/07/2023]
Abstract
Recent human studies indicate a possible correlation between the administration of antibiotics during early life and the risk of later obesity, potentially due to antibiotic-induced alteration of the gastrointestinal microbiome. In humans, the risk of obesity increases with multiple courses of antibiotics and when fetuses or infants are treated with broad-spectrum and macrolide antibiotics. In addition, the obesity risk in humans seems higher for males than females. We used a retrospective, case-control, matched-pair study design to evaluate health records for 99 control-matched pairs of rhesus macaques (Macaca mulatta) from an outdoor breeding colony. We hypothesized that NHP treated with antibiotics prior to 6 mo of age would have steeper growth curves than those who were not. However, in contrast to prior research with humans and mice, growth curves did not differ between antibiotic-treated and control animals. Differences between humans and NHP may have influenced this outcome, including the relative standardization of NHP environmental factors and diet compared with those of human populations, types of infections encountered in infancy and choice of antibiotic treatment, and the different relative maturity at 6 mo of age in the 2 species. The results provide support for current standard medical practice in NHP and highlight a difference between macaques and humans that may influence future obesity research using macaques. Determining the basis for this difference might improve our understanding of the risks of earlylife antibiotic treatment and suggest mitigation strategies for treating infant illnesses without risking obesity.
Collapse
Affiliation(s)
| | - Byung Park
- Oregon Health and Sciences University, Portland, Oregon
| | - Lina Gao
- Oregon Health and Sciences University, Portland, Oregon
| |
Collapse
|
70
|
Crowley CM, Fontaine JH, Gerriets JE, Schelegle ES, Hyde DM, Miller LA. Early life allergen and air pollutant exposures alter longitudinal blood immune profiles in infant rhesus monkeys. Toxicol Appl Pharmacol 2017; 328:60-69. [PMID: 28529118 DOI: 10.1016/j.taap.2017.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/04/2017] [Accepted: 05/09/2017] [Indexed: 10/19/2022]
Abstract
Early life is a critical period for the progressive establishment of immunity in response to environmental stimuli; the impact of airborne challenges on this process is not well defined. In a longitudinal fashion, we determined the effect of episodic house dust mite (HDM) aerosol and ozone inhalation, both separately and combined, on peripheral blood immune cell phenotypes and cytokine expression from 4 to 25weeks of age in an infant rhesus monkey model of childhood development. Immune profiles in peripheral blood were compared with lung lavage at 25weeks of age. Independent of exposure, peripheral blood cell counts fluctuated with chronologic age of animals, while IFNγ and IL-4 mRNA levels increased over time in a linear fashion. At 12weeks of age, total WBC, lymphocyte numbers, FoxP3 mRNA and IL-12 mRNA were dramatically reduced relative to earlier time points, but increased to a steady state with age. Exposure effects were observed for monocyte numbers, as well as CCR3, FoxP3, and IL-12 mRNA levels in peripheral blood. Significant differences in cell surface marker and cytokine expression were detected following in vitro HDM or PMA/ionomycin stimulation of PBMC isolated from animals exposed to either HDM or ozone. Lavage revealed a mixed immune phenotype of FoxP3, IFNγ and eosinophilia in association with combined HDM plus ozone exposure, which was not observed in blood. Collectively, our findings show that airborne challenges during postnatal development elicit measureable cell and cytokine changes in peripheral blood over time, but exposure-induced immune profiles are not mirrored in the lung.
Collapse
Affiliation(s)
- Candace M Crowley
- California National Primate Research Center, University of California, Davis, United States
| | - Justin H Fontaine
- California National Primate Research Center, University of California, Davis, United States
| | - Joan E Gerriets
- California National Primate Research Center, University of California, Davis, United States
| | - Edward S Schelegle
- California National Primate Research Center, University of California, Davis, United States; Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Dallas M Hyde
- California National Primate Research Center, University of California, Davis, United States; Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, United States; Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, United States.
| |
Collapse
|
71
|
Katagiri R, Ishihara-Hattori K, Frings W, Amano J, Fuchs A, Chiba S. Effects of SA237, a humanized anti-interleukin-6 receptor monoclonal antibody, on pre- and postnatal development in cynomolgus monkey. Birth Defects Res 2017; 109:843-856. [DOI: 10.1002/bdr2.1036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Ryuichi Katagiri
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | | | - Werner Frings
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | - Jun Amano
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| | - Antje Fuchs
- Covance Preclinical Services GmbH; Muenster Germany
| | - Shuichi Chiba
- Research Division; Chugai Pharmaceutical Co., Ltd.; Shizuoka Japan
| |
Collapse
|
72
|
Moya-Pérez A, Luczynski P, Renes IB, Wang S, Borre Y, Anthony Ryan C, Knol J, Stanton C, Dinan TG, Cryan JF. Intervention strategies for cesarean section-induced alterations in the microbiota-gut-brain axis. Nutr Rev 2017; 75:225-240. [PMID: 28379454 PMCID: PMC5410982 DOI: 10.1093/nutrit/nuw069] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial colonization of the gastrointestinal tract is an essential process that modulates host physiology and immunity. Recently, researchers have begun to understand how and when these microorganisms colonize the gut and the early-life factors that impact their natural ecological establishment. The vertical transmission of maternal microbes to the offspring is a critical factor for host immune and metabolic development. Increasing evidence also points to a role in the wiring of the gut-brain axis. This process may be altered by various factors such as mode of delivery, gestational age at birth, the use of antibiotics in early life, infant feeding, and hygiene practices. In fact, these early exposures that impact the intestinal microbiota have been associated with the development of diseases such as obesity, type 1 diabetes, asthma, allergies, and even neurodevelopmental disorders. The present review summarizes the impact of cesarean birth on the gut microbiome and the health status of the developing infant and discusses possible preventative and restorative strategies to compensate for early-life microbial perturbations.
Collapse
Affiliation(s)
- Angela Moya-Pérez
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Pauline Luczynski
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Ingrid B. Renes
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Shugui Wang
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Yuliya Borre
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - C. Anthony Ryan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Jan Knol
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Catherine Stanton
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Timothy G. Dinan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - John F. Cryan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| |
Collapse
|
73
|
Borghesi A, Stronati M, Fellay J. Neonatal Group B Streptococcal Disease in Otherwise Healthy Infants: Failure of Specific Neonatal Immune Responses. Front Immunol 2017; 8:215. [PMID: 28326082 PMCID: PMC5339282 DOI: 10.3389/fimmu.2017.00215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 12/26/2022] Open
Abstract
Only a small proportion of newborn infants exposed to a pathogenic microorganism develop overt infection. Susceptibility to infection in preterm infants and infants with known comorbidities has a likely multifactorial origin and can be often attributed to the concurrence of iatrogenic factors, environmental determinants, underlying pathogenic processes, and probably genetic predisposition. Conversely, infection occurring in otherwise healthy full-term newborn infants is unexplained in most cases. Microbial virulence factors and the unique characteristics of the neonatal immune system only partially account for the interindividual variability in the neonatal immune responses to pathogens. We here suggest that neonatal infection occurring in otherwise healthy infants is caused by a failure of the specific protective immunity to the microorganism. To explain infection in term and preterm infants, we propose an extension of the previously proposed model of the genetic architecture of infectious diseases in humans. We then focus on group B streptococcus (GBS) disease, the best characterized neonatal infection, and outline the potential molecular mechanisms underlying the selective failure of the immune responses against GBS. In light of the recent discoveries of pathogen-specific primary immunodeficiencies and of the role of anticytokine autoantibodies in increasing susceptibility to specific infections, we hypothesize that GBS disease occurring in otherwise healthy infants could reflect an immunodeficiency caused either by rare genetic defects in the infant or by transmitted maternal neutralizing antibodies. These hypotheses are consistent with available epidemiological data, with clinical and epidemiological observations, and with the state of the art of neonatal physiology and disease. Studies should now be designed to comprehensively search for genetic or immunological factors involved in susceptibility to severe neonatal infections.
Collapse
Affiliation(s)
- Alessandro Borghesi
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Mauro Stronati
- Neonatal Intensive Care Unit, San Matteo Hospital, Pavia, Italy
| | - Jacques Fellay
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
74
|
Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med 2017; 23:314-326. [PMID: 28112736 PMCID: PMC5345907 DOI: 10.1038/nm.4272] [Citation(s) in RCA: 695] [Impact Index Per Article: 86.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 12/26/2022]
Abstract
Human microbial communities are characterized by their taxonomic, metagenomic, and metabolic diversity, which varies by distinct body sites and influences human physiology. However, when and how microbial communities within each body niche acquire unique taxonomical and functional signatures in early life remains underexplored. We thus sought to assess the taxonomic composition and potential metabolic function of the neonatal and early infant microbiota across multiple body sites, and assess the impact of mode of delivery and its potential confounders or modifiers. A cohort of pregnant women in their early 3rd trimester (n=81) were prospectively enrolled for longitudinal sampling through 6 weeks post-delivery, and a second matched cross-sectional cohort (n=81) was additionally recruited for sampling once at delivery. Samples were collected for each maternal-infant dyad across multiple body sites, including stool, oral gingiva, nares, skin and vagina. 16S rRNA gene sequencing analysis and whole genome shotgun sequencing was performed to interrogate the composition and function of the neonatal and maternal microbiota. We found that the neonatal microbiota and its associated functional pathways were relatively homogenous across all body sites at delivery, with the notable exception of neonatal meconium. However, by 6 weeks, the infant microbiota structure and function had significantly expanded and diversified, with body site serving as the primary determinant of the bacterial community composition and its functional capacity. Although minor variations in the neonatal (immediately at birth) microbiota community structure were associated with Cesarean delivery in some body sites (oral, nares, and skin; R2 = 0.038), this was not true in neonatal stool (meconium, Mann-Whitney p>0.05) and there was no observable difference in community function regardless of delivery mode. By 6 weeks of age, the infant microbiota structure and function had expanded and diversified with demonstrable body site specificity (p<0.001, R2 = 0.189), and no discernable differences in neither community structure nor function by Cesarean delivery were identifiable (p=0.057, R2 = 0.007). We conclude that within the first 6 weeks of life, the infant microbiota undergoes significant reorganization that is primarily driven by body site and not by mode of delivery.
Collapse
|
75
|
Janoff EN. The microbiome and human disease pathogenesis: how do you do what you do to me …? Transl Res 2017; 179:1-6. [PMID: 27832935 DOI: 10.1016/j.trsl.2016.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 10/11/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Edward N Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), Division of Infectious Disease, Department of Medicine, University of Colorado School of Medicine, Aurora, Colo; Denver, Veterans Affairs Medical Center, Denver, Colo.
| |
Collapse
|
76
|
Gregory KE, Samuel BS, Houghteling P, Shan G, Ausubel FM, Sadreyev RI, Walker WA. Influence of maternal breast milk ingestion on acquisition of the intestinal microbiome in preterm infants. MICROBIOME 2016; 4:68. [PMID: 28034306 PMCID: PMC5200970 DOI: 10.1186/s40168-016-0214-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/29/2016] [Indexed: 05/14/2023]
Abstract
BACKGROUND The initial acquisition and early development of the intestinal microbiome during infancy are important to human health across the lifespan. Mode of birth, antibiotic administration, environment of care, and nutrition have all been shown to play a role in the assembly of the intestinal microbiome during early life. For preterm infants, who are disproportionately at risk of inflammatory intestinal disease (i.e., necrotizing enterocolitis), a unique set of clinical factors influence the establishment of the microbiome. The purpose of this study was to establish the influence of nutritional exposures on the intestinal microbiome in a cohort of preterm infants early in life. RESULTS Principal component analysis of 199 samples from 30 preterm infants (<32 weeks) over the first 60 days following birth showed that the intestinal microbiome was influenced by postnatal time (p < 0.001, R 2 = 0.13), birth weight (p < 0.001, R 2 = 0.08), and nutrition (p < 0.001, R 2 = 0.21). Infants who were fed breast milk had a greater initial bacterial diversity and a more gradual acquisition of diversity compared to infants who were fed infant formula. The microbiome of infants fed breast milk were more similar regardless of birth weight (p = 0.049), in contrast to the microbiome of infants fed infant formula, which clustered differently based on birth weight (p < 0.001). By adjusting for differences in gut maturity, an ordered succession of microbial phylotypes was observed in breast milk-fed infants, which appeared to be disrupted in those fed infant formula. Supplementation with pasteurized donor human milk was partially successful in promoting a microbiome more similar to breast milk-fed infants and moderating rapid increases in bacterial diversity. CONCLUSIONS The preterm infant intestinal microbiome is influenced by postnatal time, birth weight, gestational age, and nutrition. Feeding with breast milk appears to mask the influence of birth weight, suggesting a protective effect against gut immaturity in the preterm infant. These findings suggest not only a microbial mechanism underpinning the body of evidence showing that breast milk promotes intestinal health in the preterm infant but also a dynamic interplay of host and dietary factors that facilitate the colonization of and enrichment for specific microbes during establishment of the preterm infant microbiota.
Collapse
Affiliation(s)
- Katherine E. Gregory
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA
| | - Buck S. Samuel
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030 USA
| | - Pearl Houghteling
- Department of Pediatrics, Yale School of Medicine, New Haven, CT USA
| | - Guru Shan
- Cooper Medical School, Camden, NJ USA
| | - Frederick M. Ausubel
- Department of Molecular Biology, Massachusetts General Hospital, Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, Harvard Medical School, Boston, MA USA
| | - W. Allan Walker
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA USA
| |
Collapse
|
77
|
The Impact of the Gut Microbiota on Humoral Immunity to Pathogens and Vaccination in Early Infancy. PLoS Pathog 2016; 12:e1005997. [PMID: 28006021 PMCID: PMC5179050 DOI: 10.1371/journal.ppat.1005997] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
78
|
Kabagambe EK, Ezeamama AE, Guwatudde D, Campos H, Fawzi W. Plasma n-6 Fatty Acid Levels Are Associated With CD4 Cell Counts, Hospitalization, and Mortality in HIV-Infected Patients. J Acquir Immune Defic Syndr 2016; 73:598-605. [PMID: 27529612 PMCID: PMC5110389 DOI: 10.1097/qai.0000000000001149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Fatty acids, including n-6 series, modulate immune function, but their effect on CD4 cell counts, death, or hospitalization in HIV-infected patients on antiretroviral therapy is unknown. METHODS In a randomized trial for effects of multivitamins in HIV-infected patients in Uganda, we used gas chromatography to measure plasma n-6 fatty acids at baseline; determined CD4 counts at baseline, 3, 6, 12, and 18 months; and recorded hospitalization or death events. The associations of fatty acids with CD4 counts and events were analyzed using repeated-measures analysis of variance and Cox regression, respectively. RESULTS Among 297 patients with fatty acids measurements, 16 patients died and 69 were hospitalized within 18 months. Except for linoleic acid, n-6 fatty acids levels were positively associated with CD4 counts at baseline but not during follow-up. In models that included all 5 major n-6 fatty acids, age; sex; body mass index; anemia status; use of antiretroviral therapy, multivitamin supplements, and alcohol; and the risk of death or hospitalization decreased significantly with an increase in linoleic acid and gamma-linolenic acid levels, whereas associations for dihomo-gamma-linolenic acid, arachidonic acid, and aolrenic acid were null. The hazard ratios (95% confidence intervals) per 1 SD increase in linoleic acid and gamma-linolenic acid were 0.73 (0.56-0.94) and 0.51 (0.36-0.72), respectively. Gamma-linolenic acid remained significant (hazard ratio = 0.51; 95% confidence interval: 0.35 to 0.68) after further adjustment for other plasma fatty acids. CONCLUSIONS Lower levels of gamma-linolenic acid are associated with lower CD4 counts and an increased risk of death or hospitalization. These results suggest a potential for using n-6 fatty acids to improve outcomes from antiretroviral therapy.
Collapse
Affiliation(s)
- Edmond K Kabagambe
- Division of Epidemiology, Department of Medicine and the Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37203
| | - Amara E Ezeamama
- Department of Epidemiology and Biostatistics, The University of Georgia, Athens, GA 30602
| | - David Guwatudde
- Department of Epidemiology and Biostatistics (DG), School of Public Health, Makerere University College of Health Sciences, P.O. Box 7072, Kampala, Uganda
| | - Hannia Campos
- Department of Nutrition and Departments of Global Health and Population and Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115
| | - Wafaie Fawzi
- Department of Nutrition and Departments of Global Health and Population and Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115
| |
Collapse
|
79
|
Kohl KD, Brun A, Magallanes M, Brinkerhoff J, Laspiur A, Acosta JC, Caviedes-Vidal E, Bordenstein SR. Gut microbial ecology of lizards: insights into diversity in the wild, effects of captivity, variation across gut regions and transmission. Mol Ecol 2016; 26:1175-1189. [PMID: 27862531 DOI: 10.1111/mec.13921] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/13/2022]
Abstract
Animals maintain complex associations with a diverse microbiota living in their guts. Our understanding of the ecology of these associations is extremely limited in reptiles. Here, we report an in-depth study into the microbial ecology of gut communities in three syntopic and viviparous lizard species (two omnivores: Liolaemus parvus and Liolaemus ruibali and an herbivore: Phymaturus williamsi). Using 16S rRNA gene sequencing to inventory various bacterial communities, we elucidate four major findings: (i) closely related lizard species harbour distinct gut bacterial microbiota that remain distinguishable in captivity; a considerable portion of gut bacterial diversity (39.1%) in nature overlap with that found on plant material, (ii) captivity changes bacterial community composition, although host-specific communities are retained, (iii) faecal samples are largely representative of the hindgut bacterial community and thus represent acceptable sources for nondestructive sampling, and (iv) lizards born in captivity and separated from their mothers within 24 h shared 34.3% of their gut bacterial diversity with their mothers, suggestive of maternal or environmental transmission. Each of these findings represents the first time such a topic has been investigated in lizard hosts. Taken together, our findings provide a foundation for comparative analyses of the faecal and gastrointestinal microbiota of reptile hosts.
Collapse
Affiliation(s)
- Kevin D Kohl
- Department of Biological Sciences, Vanderbilt University, 465 21st Ave South, Nashville, TN, 37235, USA.,Laboratorio de Biología Integrativa, Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas, San Luis, 5700, Argentina.,Departamento de Bioquímica y Ciencias Biológicas, Universidad Nacional de San Luis, Chacabuco 917, San Luis, 5700, Argentina
| | - Antonio Brun
- Laboratorio de Biología Integrativa, Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas, San Luis, 5700, Argentina.,Departamento de Bioquímica y Ciencias Biológicas, Universidad Nacional de San Luis, Chacabuco 917, San Luis, 5700, Argentina
| | - Melisa Magallanes
- Laboratorio de Biología Integrativa, Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas, San Luis, 5700, Argentina.,Departamento de Bioquímica y Ciencias Biológicas, Universidad Nacional de San Luis, Chacabuco 917, San Luis, 5700, Argentina
| | - Joshua Brinkerhoff
- Laboratorio de Biología Integrativa, Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas, San Luis, 5700, Argentina.,Departamento de Bioquímica y Ciencias Biológicas, Universidad Nacional de San Luis, Chacabuco 917, San Luis, 5700, Argentina
| | - Alejandro Laspiur
- Centro de Investigaciones de la Geósfera y la Biósfera (CIGEOBIO-CONICET) - Departamento de Biología, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de San Juan, Av. José I. de la Roza 590 Oeste, J5402DCS, San Juan, Argentina
| | - Juan Carlos Acosta
- Centro de Investigaciones de la Geósfera y la Biósfera (CIGEOBIO-CONICET) - Departamento de Biología, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de San Juan, Av. José I. de la Roza 590 Oeste, J5402DCS, San Juan, Argentina
| | - Enrique Caviedes-Vidal
- Laboratorio de Biología Integrativa, Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, Consejo Nacional de Investigaciones Científicas y Técnicas, San Luis, 5700, Argentina.,Departamento de Bioquímica y Ciencias Biológicas, Universidad Nacional de San Luis, Chacabuco 917, San Luis, 5700, Argentina
| | - Seth R Bordenstein
- Department of Biological Sciences, Vanderbilt University, 465 21st Ave South, Nashville, TN, 37235, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University, 465 21st Ave South, Nashville, TN, 37235, USA
| |
Collapse
|
80
|
Sutherland DB, Suzuki K, Fagarasan S. Fostering of advanced mutualism with gut microbiota by Immunoglobulin A. Immunol Rev 2016; 270:20-31. [PMID: 26864102 DOI: 10.1111/imr.12384] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunoglobulin A (IgA), the most abundantly secreted antibody isotype in mammals, not only provides direct immune protection to neonates via maternal milk but also helps program the infant immune system by regulating the microbiota. IgA continues to maintain dynamic interactions with the gut microbiota throughout life and this influences immune system homeostasis as well as other physiological processes. The secretory IgA produced independently of T-cell selection are commonly referred to as natural or innate antibodies. Our studies have shown that innate-IgA, while effective at excluding microorganisms from the gut, does not promote mutualism with the microbiota in the same way as adaptive-IgA that is selected in T cell-dependent germinal center reactions. Adaptive-IgA fosters more advanced mutualism with the microbiota than innate-IgA by selecting and diversifying beneficial microbial communities. In this review, we suggest that the diversified microbiota resulting from adaptive-IgA pressure was pivotal in promoting ecological adaptability and speciation potential of mammals.
Collapse
Affiliation(s)
- Duncan B Sutherland
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Keiichiro Suzuki
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences (IMS), RIKEN Yokohama Institute, Yokohama, Japan
| |
Collapse
|
81
|
Dietary Gluten-Induced Gut Dysbiosis Is Accompanied by Selective Upregulation of microRNAs with Intestinal Tight Junction and Bacteria-Binding Motifs in Rhesus Macaque Model of Celiac Disease. Nutrients 2016; 8:nu8110684. [PMID: 27801835 PMCID: PMC5133072 DOI: 10.3390/nu8110684] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022] Open
Abstract
The composition of the gut microbiome reflects the overall health status of the host. In this study, stool samples representing the gut microbiomes from 6 gluten-sensitive (GS) captive juvenile rhesus macaques were compared with those from 6 healthy, age- and diet-matched peers. A total of 48 samples representing both groups were studied using V4 16S rRNA gene DNA analysis. Samples from GS macaques were further characterized based on type of diet administered: conventional monkey chow, i.e., wheat gluten-containing diet (GD), gluten-free diet (GFD), barley gluten-derived diet (BOMI) and reduced gluten barley-derived diet (RGB). It was hypothesized that the GD diet would lower the gut microbial diversity in GS macaques. This is the first report illustrating the reduction of gut microbial alpha-diversity (p < 0.05) following the consumption of dietary gluten in GS macaques. Selected bacterial families (e.g., Streptococcaceae and Lactobacillaceae) were enriched in GS macaques while Coriobacteriaceae was enriched in healthy animals. Within several weeks after the replacement of the GD by the GFD diet, the composition (beta-diversity) of gut microbiome in GS macaques started to change (p = 0.011) towards that of a normal macaque. Significance for alpha-diversity however, was not reached by the day 70 when the feeding experiment ended. Several inflammation-associated microRNAs (miR-203, -204, -23a, -23b and -29b) were upregulated (p < 0.05) in jejunum of 4 biopsied GS macaques fed GD with predicted binding sites on 16S ribosomal RNA of Lactobacillus reuteri (accession number: NR_025911), Prevotella stercorea (NR_041364) and Streptococcus luteciae (AJ297218) that were overrepresented in feces. Additionally, claudin-1, a validated tight junction protein target of miR-29b was significantly downregulated in jejunal epithelium of GS macaques. Taken together, we predict that with the introduction of effective treatments in future studies the diversity of gut microbiomes in GS macaques will approach those of healthy individuals. Further studies are needed to elucidate the regulatory pathways of inflammatory miRNAs in intestinal mucosa of GS macaques and to correlate their expression with gut dysbiosis.
Collapse
|
82
|
Janoff EN. WITHDRAWN: The microbiome and human disease pathogenesis: how do you do what you do to me …? Transl Res 2016:S1931-5244(16)30273-0. [PMID: 27832937 DOI: 10.1016/j.trsl.2016.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/10/2016] [Indexed: 11/30/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Edward N Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colo; Denver, Veterans Affairs Medical Center, Denver, Colo
| |
Collapse
|
83
|
Davis MY, Zhang H, Brannan LE, Carman RJ, Boone JH. Rapid change of fecal microbiome and disappearance of Clostridium difficile in a colonized infant after transition from breast milk to cow milk. MICROBIOME 2016; 4:53. [PMID: 27717398 PMCID: PMC5055705 DOI: 10.1186/s40168-016-0198-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/23/2016] [Indexed: 05/12/2023]
Abstract
BACKGROUND Clostridium difficile is the most common known cause of antibiotic-associated diarrhea. Upon the disturbance of gut microbiota by antibiotics, C. difficile establishes growth and releases toxins A and B, which cause tissue damage in the host. The symptoms of C. difficile infection disease range from mild diarrhea to pseudomembranous colitis and toxic megacolon. Interestingly, 10-50 % of infants are asymptomatic carriers of C. difficile. This longitudinal study of the C. difficile colonization in an infant revealed the dynamics of C. difficile presence in gut microbiota. METHODS Fifty fecal samples, collected weekly between 5.5 and 17 months of age from a female infant who was an asymptomatic carrier of C. difficile, were analyzed by 16S rRNA gene sequencing. RESULTS Colonization switching between toxigenic and non-toxigenic C. difficile strains as well as more than 100,000-fold fluctuations of C. difficile counts were observed. C. difficile toxins were detected during the testing period in some infant stool samples, but the infant never had diarrhea. Although fecal microbiota was stable during breast feeding, a dramatic and permanent change of microbiota composition was observed within 5 days of the transition from human milk to cow milk. A rapid decline and eventual disappearance of C. difficile coincided with weaning at 12.5 months. An increase in the relative abundance of Bacteroides spp., Blautia spp., Parabacteroides spp., Coprococcus spp., Ruminococcus spp., and Oscillospira spp. and a decrease of Bifidobacterium spp., Lactobacillus spp., Escherichia spp., and Clostridium spp. were observed during weaning. The change in microbiome composition was accompanied by a gradual increase of fecal pH from 5.5 to 7. CONCLUSIONS The bacterial groups that are less abundant in early infancy, and that increase in relative abundance after weaning, likely are responsible for the expulsion of C. difficile.
Collapse
Affiliation(s)
- Manli Y Davis
- TechLab, Inc., 2001 Kraft Drive, Blacksburg, VA, 24060, USA.
| | - Husen Zhang
- Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Lera E Brannan
- TechLab, Inc., 2001 Kraft Drive, Blacksburg, VA, 24060, USA
| | | | - James H Boone
- TechLab, Inc., 2001 Kraft Drive, Blacksburg, VA, 24060, USA.
| |
Collapse
|
84
|
Chu DM, Meyer KM, Prince AL, Aagaard KM. Impact of maternal nutrition in pregnancy and lactation on offspring gut microbial composition and function. Gut Microbes 2016; 7:459-470. [PMID: 27686144 PMCID: PMC5103658 DOI: 10.1080/19490976.2016.1241357] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Evidence supporting the Developmental Origins of Health and Disease Hypothesis indicates that maternal nutrition in pregnancy has a significant impact on offspring disease risk later in life, likely by modulating developmental processes in utero. Gut microbiota have recently been explored as a potential mediating factor, as dietary components strongly influence microbiota abundance, function and its impact on host physiology. A growing body of evidence has additionally indicated that the intrauterine environment is not sterile as once presumed, indicating that maternal-fetal transmission of microbiota may occur during pregnancy. In this article, we will review the body of literature that supports this emerging hypothesis, as well as highlight the work in relevant animal models demonstrating associations with maternal gestational nutrition and the offspring gut microbiome that may influence offspring physiology and susceptibility to disease.
Collapse
Affiliation(s)
- Derrick M. Chu
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Kristen M. Meyer
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX,Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX
| | - Amanda L. Prince
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX,Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX,Department of Molecular & Cell Biology, Baylor College of Medicine, Houston, TX,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX,CONTACT Kjersti M. Aagaard, MD PhD FACOG Associate Professor, Baylor College of Medicine, Division of Maternal-Fetal Medicine, One Baylor Plaza, Jones 314, Houston, TX, 77030
| |
Collapse
|
85
|
Jeffery HC, Braitch MK, Brown S, Oo YH. Clinical Potential of Regulatory T Cell Therapy in Liver Diseases: An Overview and Current Perspectives. Front Immunol 2016; 7:334. [PMID: 27656181 PMCID: PMC5012133 DOI: 10.3389/fimmu.2016.00334] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/19/2016] [Indexed: 01/06/2023] Open
Abstract
The increasing demand for liver transplantation and the decline in donor organs has highlighted the need for alternative novel therapies to prevent chronic active hepatitis, which eventually leads to liver cirrhosis and liver cancer. Liver histology of chronic hepatitis is composed of both effector and regulatory lymphocytes. The human liver contains different subsets of effector lymphocytes that are kept in check by a subpopulation of T cells known as Regulatory T cells (Treg). The balance of effector and regulatory lymphocytes generally determines the outcome of hepatic inflammation: resolution, fulminant hepatitis, or chronic active hepatitis. Thus, maintaining and adjusting this balance is crucial in immunological manipulation of liver diseases. One of the options to restore this balance is to enrich Treg in the liver disease patients. Advances in the knowledge of Treg biology and development of clinical grade isolation reagents, cell sorting equipment, and good manufacturing practice facilities have paved the way to apply Treg cells as a potential therapy to restore peripheral self-tolerance in autoimmune liver diseases (AILD), chronic rejection, and posttransplantation. Past and on-going studies have applied Treg in type-1 diabetes mellitus, systemic lupus erythematosus, graft versus host diseases, and solid organ transplantations. There have not been any new therapies for the AILD for more than three decades; thus, the clinical potential for the application of autologous Treg cell therapy to treat autoimmune liver disease is an attractive and novel option. However, it is fundamental to understand the deep immunology, genetic profiles, biology, homing behavior, and microenvironment of Treg before applying the cells to the patients.
Collapse
Affiliation(s)
- Hannah C Jeffery
- NIHR Biomedical Research Unit in Liver Diseases, Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Manjit Kaur Braitch
- NIHR Biomedical Research Unit in Liver Diseases, Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Solomon Brown
- NIHR Biomedical Research Unit in Liver Diseases, Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Ye Htun Oo
- NIHR Biomedical Research Unit in Liver Diseases, Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; Liver and Hepatobiliary Unit, University Hospital NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
86
|
Zhou S, Xu R, He F, Zhou J, Wang Y, Zhou J, Wang M, Zhou W. Diversity of Gut Microbiota Metabolic Pathways in 10 Pairs of Chinese Infant Twins. PLoS One 2016; 11:e0161627. [PMID: 27583441 PMCID: PMC5008625 DOI: 10.1371/journal.pone.0161627] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Early colonization of gut microbiota in human gut is a complex process. It remains unclear when gut microbiota colonization occurs and how it proceeds. In order to study gut microbiota composition in human early life, the present study recruited 10 healthy pairs of twins, including five monozygotic (MZ) and five dizygotic (DZ) twin pairs, whose age ranged from 0 to 6 years old. 20 fecal samples from these twins were processed by shotgun metagenomic sequencing, and their averaged data outputs were generated as 2G per sample. We used MEGAN5 to perform taxonomic and functional annotation of the metagenomic data, and systematically analyzed those 20 samples, including Jaccard index similarity, principle component, clustering, and correlation analyses. Our findings indicated that within our study group: 1) MZ-twins share more microbes than DZ twins or non-twin pairs, 2) gut microbiota distribution is relatively stable at metabolic pathways level, 3) age represents the strongest factor that can account for variation in gut microbiota, and 4) a clear metabolic pathway shift can be observed, which speculatively occurs around the age of 1 year old. This research will serve as a base for future studies of gut microbiota-related disease research.
Collapse
Affiliation(s)
- Shaoming Zhou
- Division of Gastroenterology, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
- * E-mail: (MW); (SZ); (WZ)
| | - Ruihuan Xu
- Clinical Laboratory, Longgang Central Hospital of Shenzhen, Guangdong, China
| | - Fusheng He
- Shenzhen Following Precision Medical Research Institute, Shenzhen, Guangdong, China
| | - Jiaxiu Zhou
- Division of psychology, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Yan Wang
- Shenzhen Imuno Biotech Co.,Ltd, Shenzhen, China
| | - Jianli Zhou
- Division of Gastroenterology, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Mingbang Wang
- Shenzhen Following Precision Medical Research Institute, Shenzhen, Guangdong, China
- Shenzhen Imuno Biotech Co.,Ltd, Shenzhen, China
- Division of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China
- * E-mail: (MW); (SZ); (WZ)
| | - Wenhao Zhou
- Division of Neonatology, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Birth Defects, Children’s Hospital of Fudan University, Shanghai, China
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China
- * E-mail: (MW); (SZ); (WZ)
| |
Collapse
|
87
|
Chu DM, Antony KM, Ma J, Prince AL, Showalter L, Moller M, Aagaard KM. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med 2016; 8:77. [PMID: 27503374 PMCID: PMC4977686 DOI: 10.1186/s13073-016-0330-z] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/01/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Emerging evidence suggests that the in utero environment is not sterile as once presumed. Work in the mouse demonstrated transmission of commensal bacteria from mother to fetus during gestation, though it is unclear what modulates this process. We have previously shown in the nonhuman primate that, independent of obesity, a maternal high-fat diet during gestation and lactation persistently shapes the juvenile gut microbiome. We therefore sought to interrogate in a population-based human longitudinal cohort whether a maternal high-fat diet similarly alters the neonatal and infant gut microbiome in early life. METHODS A representative cohort was prospectively enrolled either in the early third trimester or intrapartum (n = 163), with a subset consented to longitudinal sampling through the postpartum interval (n = 81). Multiple body site samples, including stool and meconium, were collected from neonates at delivery and by 6 weeks of age. A rapid dietary questionnaire was administered to estimate intake of fat, added sugars, and fiber over the past month (National Health and Examination Survey). DNA was extracted from each infant meconium/stool sample (MoBio) and subjected to 16S rRNA gene sequencing and analysis. RESULTS On average, the maternal dietary intake of fat ranged from 14.0 to 55.2 %, with an average intake of 33.1 % (σ = 6.1 %). Mothers whose diets significantly differed from the mean (±1 standard deviation) were separated into two distinct groups, a control group (n = 13, μ = 24.4 %) and a high-fat group (n = 13, μ = 43.1 %). Principal coordinate analysis revealed that the microbiome of the neonatal stool at birth (meconium) clustered differently by virtue of maternal gestational diet (PERMANOVA p = 0.001). LEfSe feature selection identified several taxa that discriminated the groups, with a notable relative depletion of Bacteroides in the neonates exposed to a maternal high-fat gestational diet (Student's t-test, p < 0.05) that persisted to 6 weeks of age. CONCLUSIONS Similar to the primate, independent of maternal body mass index, a maternal high-fat diet is associated with distinct changes in the neonatal gut microbiome at birth which persist through 4-6 weeks of age. Our findings underscore the importance of counseling pregnant mothers on macronutrient consumption during pregnancy and lactation.
Collapse
Affiliation(s)
- Derrick M Chu
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Kathleen M Antony
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Jun Ma
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Amanda L Prince
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Lori Showalter
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Michelle Moller
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
- Departments of Molecular & Human Genetics, Molecular & Cell Biology, and Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.
- Division of Maternal-Fetal Medicine, Baylor College of Medicine, One Baylor Plaza, Jones 314, Houston, TX, 77030, USA.
| |
Collapse
|
88
|
Montoya J, Matta NB, Suchon P, Guzian MC, Lambert NC, Mattei JP, Guis S, Breban M, Roudier J, Balandraud N. Patients with ankylosing spondylitis have been breast fed less often than healthy controls: a case-control retrospective study. Ann Rheum Dis 2016; 75:879-82. [PMID: 26458738 DOI: 10.1136/annrheumdis-2015-208187] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/20/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) is a chronic inflammatory disease affecting the spine and pelvis of young adults. On the HLA-B27 genetic background, the occurrence of AS is influenced by the intestinal microbiota. The goal of our study was to test whether breast feeding, which influences microbiota, can prevent the development of AS. METHODS First, 203 patients with HLA-B27-positive AS fulfilling the modified New York criteria were recruited in the Department of Rheumatology, Ste Marguerite hospital in Marseilles. A total of 293 healthy siblings were also recruited to make up a control group within the same families. Second, 280 healthy controls, and 100 patients with rheumatoid arthritis and their siblings were recruited. The data collected were age, gender, number of brothers and sisters, age at disease onset, type and duration of feeding (breast or bottle). RESULTS Patients with AS had been breast fed less often than healthy controls. In families where children were breast fed, the patients with AS were less often breast fed than their healthy siblings (57% vs 72%), giving an OR for AS onset of 0.53 (95% CI (0.36 to 0.77), p value=0.0009). Breast feeding reduced familial prevalence of AS. The frequency of breast feeding was similar in the AS siblings and in the 280 unrelated controls. However, patients with AS were less often breast fed compared with the 280 unrelated controls (OR 0.6, 95% CI (0.42 to 0.89), p<0.01). CONCLUSIONS Our study suggests a breastfeeding-induced protective effect on the occurrence of AS. To our knowledge, this is the first study of breastfeeding history in patients with AS.
Collapse
Affiliation(s)
- J Montoya
- Department of Rheumatology, IML, AP-HM, Marseille, France
| | - N B Matta
- Department of Rheumatology, IML, AP-HM, Marseille, France
| | - P Suchon
- Departments of Nutrition, Obesity and Risk of Thrombosis, INSERM, UMR1062, Aix-Marseille University, Marseille, France Laboratory of Hematology, AP-HM, Hopital Timone, Marseille, France
| | - M C Guzian
- Department of Rheumatology, IML, AP-HM, Marseille, France
| | - N C Lambert
- INSERM UMRs 1097, Aix Marseille Université, Marseille, France
| | - J P Mattei
- Department of Rheumatology, IML, AP-HM, Marseille, France
| | - S Guis
- Department of Rheumatology, IML, AP-HM, Marseille, France
| | - M Breban
- INSERM U987 and Université de Versailles St.-Quentin-en-Yvelines, Montigny-le-Bretonneux, France Hôpital Ambroise Paré, AP-HP, and Université de Versailles St.-Quentin-en-Yvelines, Boulogne-Billancourt, France Laboratoire d'Excellence Inflamex, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - J Roudier
- Department of Rheumatology, IML, AP-HM, Marseille, France INSERM UMRs 1097, Aix Marseille Université, Marseille, France
| | - N Balandraud
- Department of Rheumatology, IML, AP-HM, Marseille, France INSERM UMRs 1097, Aix Marseille Université, Marseille, France
| |
Collapse
|
89
|
Vatanen T, Kostic AD, d'Hennezel E, Siljander H, Franzosa EA, Yassour M, Kolde R, Vlamakis H, Arthur TD, Hämäläinen AM, Peet A, Tillmann V, Uibo R, Mokurov S, Dorshakova N, Ilonen J, Virtanen SM, Szabo SJ, Porter JA, Lähdesmäki H, Huttenhower C, Gevers D, Cullen TW, Knip M, Xavier RJ. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016; 165:842-53. [PMID: 27133167 DOI: 10.1016/j.cell.2016.04.007] [Citation(s) in RCA: 781] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/19/2016] [Accepted: 03/31/2016] [Indexed: 12/21/2022]
Abstract
According to the hygiene hypothesis, the increasing incidence of autoimmune diseases in western countries may be explained by changes in early microbial exposure, leading to altered immune maturation. We followed gut microbiome development from birth until age three in 222 infants in Northern Europe, where early-onset autoimmune diseases are common in Finland and Estonia but are less prevalent in Russia. We found that Bacteroides species are lowly abundant in Russians but dominate in Finnish and Estonian infants. Therefore, their lipopolysaccharide (LPS) exposures arose primarily from Bacteroides rather than from Escherichia coli, which is a potent innate immune activator. We show that Bacteroides LPS is structurally distinct from E. coli LPS and inhibits innate immune signaling and endotoxin tolerance; furthermore, unlike LPS from E. coli, B. dorei LPS does not decrease incidence of autoimmune diabetes in non-obese diabetic mice. Early colonization by immunologically silencing microbiota may thus preclude aspects of immune education.
Collapse
Affiliation(s)
- Tommi Vatanen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Computer Science, Aalto University School of Science, 02150 Espoo, Finland
| | - Aleksandar D Kostic
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Eva d'Hennezel
- Novartis Institutes for Biomedical Research Inc., Cambridge, MA 02139, USA
| | - Heli Siljander
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland; Research Programs Unit, Diabetes and Obesity Research Program, University of Helsinki, 00290 Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland
| | - Eric A Franzosa
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Moran Yassour
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Raivo Kolde
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Anu-Maaria Hämäläinen
- Department of Pediatrics, Jorvi Hospital, Helsinki University Hospital, 02740 Espoo, Finland
| | - Aleksandr Peet
- Department of Pediatrics, University of Tartu and Tartu University Hospital, 51014 Tartu, Estonia
| | - Vallo Tillmann
- Department of Pediatrics, University of Tartu and Tartu University Hospital, 51014 Tartu, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Sergei Mokurov
- Ministry of Health and Social Development, Karelian Republic of the Russian Federation, Lenin Street 6, 185035 Petrozavodsk, Russia
| | - Natalya Dorshakova
- Petrozavodsk State University, Department of Family Medicine, Lenin Street 33, 185910 Petrozavodsk, Russia
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, 20520 Turku, Finland; Department of Clinical Microbiology, University of Eastern Finland, 70211 Kuopio, Finland
| | - Suvi M Virtanen
- Department of Health, National Institute for Health and Welfare, 00271 Helsinki, Finland; School of Health Sciences, University of Tampere, 33014 Tampere, Finland; Science Centre, Pirkanmaa Hospital District and Research Center for Child Health, University Hospital, 33521 Tampere, Finland
| | - Susanne J Szabo
- Novartis Institutes for Biomedical Research Inc., Cambridge, MA 02139, USA
| | - Jeffrey A Porter
- Novartis Institutes for Biomedical Research Inc., Cambridge, MA 02139, USA
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University School of Science, 02150 Espoo, Finland
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA
| | - Dirk Gevers
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas W Cullen
- Novartis Institutes for Biomedical Research Inc., Cambridge, MA 02139, USA
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland; Research Programs Unit, Diabetes and Obesity Research Program, University of Helsinki, 00290 Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland; Folkhälsan Research Center, 00290 Helsinki, Finland
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
90
|
Yeruva L, Spencer NE, Saraf MK, Hennings L, Bowlin AK, Cleves MA, Mercer K, Chintapalli SV, Shankar K, Rank RG, Badger TM, Ronis MJJ. Formula diet alters small intestine morphology, microbial abundance and reduces VE-cadherin and IL-10 expression in neonatal porcine model. BMC Gastroenterol 2016; 16:40. [PMID: 27005303 PMCID: PMC4804644 DOI: 10.1186/s12876-016-0456-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/15/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Breastfeeding is associated with a variety of positive health outcomes in children and is recommended exclusively for the first 6 months of life; however, 50-70 % of infants in the US are formula-fed. To test the hypothesis that immune system development and function in neonates and infants are significantly influenced by diet, 2-day old piglets were fed soy or milk formula (n = 6/group/gender) until day 21 and compared to a sow-fed group (n = 6/gender). METHODS Histomorphometric analyses of ileum, jejunum and Peyer's patches were carried out, to determine the inflammation status, mRNA and protein expression of pro-inflammatory, anti-inflammatory and growth-related chemokines and cytokines. RESULTS In formula-fed animals, increases in ileum and jejunum villus height and crypt depth were observed in comparison to sow-fed animals (jejunum, p < 0.01 villus height, p < 0.04 crypt depth; ileum p < 0.001 villus height, p < 0.002 crypt depth). In formula-fed the lymphoid follicle size (p < 0.01) and germinal centers (p < 0.01) with in the Peyer's patch were significantly decreased in comparison to sow-fed, indicating less immune education. In ileum, formula diet induced significant up-regulation of AMCFII, IL-8, IL-15, VEGFA, LIF, FASL, CXCL11, CCL4, CCL25 and down-regulation of IL-6, IL-9, IL-10, IL-27, IFNA4, CSF3, LOC100152038, and LOC100736831 at the transcript level. We have confirmed some of the mRNA data by measuring protein, and significant down-regulation of anti-inflammatory molecule IL-10 in comparison to sow-fed piglets was observed. To further determine the membrane protein expression in the ileum, VE-cadherin, occludin, and claudin-3, Western blot analyses were conducted. Sow fed piglets showed significantly more VE-Cadherin, which associated with levels of calcium, and putrescine measured. It is possible that differences in GI tract and immune development are related to shifts in the microbiome; notably, there were 5-fold higher amounts of Lactobacillaceae spp and 3 fold higher Clostridia spp in the sow fed group in comparison to milk formula-fed piglets, whereas in milk formula-fed pigs Enterobacteriaceae spp was 5-fold higher. CONCLUSION In conclusion, formula diet alters GI morphology, microbial abundance, intestinal barrier protein VE-cadherin and anti-inflammatory molecule IL-10 expression. Further characterization of formula effects could lead to modification of infant formula to improve immune function, reduce inflammation and prevent conditions such as allergies and infections.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Calcium/metabolism
- Cytokines/drug effects
- Cytokines/genetics
- Cytokines/metabolism
- Diet
- Down-Regulation
- Fas Ligand Protein/drug effects
- Fas Ligand Protein/genetics
- Fas Ligand Protein/metabolism
- Gastrointestinal Microbiome/drug effects
- Humans
- Ileum/drug effects
- Ileum/metabolism
- Ileum/microbiology
- Ileum/pathology
- Infant Formula/pharmacology
- Infant, Newborn
- Interferon-alpha/drug effects
- Interferon-alpha/genetics
- Interferon-alpha/metabolism
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Interleukin-15/genetics
- Interleukin-15/metabolism
- Interleukin-27/genetics
- Interleukin-27/metabolism
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Interleukin-8/drug effects
- Interleukin-8/genetics
- Interleukin-8/metabolism
- Interleukin-9/genetics
- Interleukin-9/metabolism
- Intestine, Small/drug effects
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Intestine, Small/pathology
- Jejunum/drug effects
- Jejunum/metabolism
- Jejunum/microbiology
- Jejunum/pathology
- Leukemia Inhibitory Factor/drug effects
- Leukemia Inhibitory Factor/genetics
- Leukemia Inhibitory Factor/metabolism
- Milk
- Peyer's Patches/drug effects
- Peyer's Patches/immunology
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Soy Foods
- Swine
- Up-Regulation
- Vascular Endothelial Growth Factor A/drug effects
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Laxmi Yeruva
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Arkansas Children’s Hospital Research Institute, Little Rock, USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | | | - Manish K. Saraf
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Leah Hennings
- />Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Anne K. Bowlin
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Mario A. Cleves
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Kelly Mercer
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Sree V. Chintapalli
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Kartik Shankar
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Roger G. Rank
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Thomas M. Badger
- />Arkansas Children’s Nutrition Center, 15 Children’s Way, Little Rock, AR 72202 USA
- />Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, USA
| | - Martin J. J. Ronis
- />Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA USA
| |
Collapse
|
91
|
Abstract
Ecological developmental biology is the study of the interactions between developing organisms and their environments. Organisms have evolved to use the environment as a source of important cues that can alter the trajectory of their development. First, developmental plasticity enables the genome to generate a repertoire of possible phenotypes, and environmental cues are often used to select the phenotype that appears most adaptive at that time. This facilitates evolutionary strategies such as phenotypic accommodation, genetic assimilation, and niche construction. Second, developmental symbiosis, wherein the developing animal utilizes cues from other organisms for normal cell differentiation and morphogenesis, has been found to be ubiquitous. The coevolution of symbiotic microbes and animal cells has often led to the dependency of an animal's development on particular microbial signals, making these cues essential and expected components of normal development.
Collapse
|
92
|
Victora CG, Bahl R, Barros AJD, França GVA, Horton S, Krasevec J, Murch S, Sankar MJ, Walker N, Rollins NC. Breastfeeding in the 21st century: epidemiology, mechanisms, and lifelong effect. Lancet 2016; 387:475-90. [PMID: 26869575 DOI: 10.1016/s0140-6736(15)01024-7] [Citation(s) in RCA: 4016] [Impact Index Per Article: 446.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The importance of breastfeeding in low-income and middle-income countries is well recognised, but less consensus exists about its importance in high-income countries. In low-income and middle-income countries, only 37% of children younger than 6 months of age are exclusively breastfed. With few exceptions, breastfeeding duration is shorter in high-income countries than in those that are resource-poor. Our meta-analyses indicate protection against child infections and malocclusion, increases in intelligence, and probable reductions in overweight and diabetes. We did not find associations with allergic disorders such as asthma or with blood pressure or cholesterol, and we noted an increase in tooth decay with longer periods of breastfeeding. For nursing women, breastfeeding gave protection against breast cancer and it improved birth spacing, and it might also protect against ovarian cancer and type 2 diabetes. The scaling up of breastfeeding to a near universal level could prevent 823,000 annual deaths in children younger than 5 years and 20,000 annual deaths from breast cancer. Recent epidemiological and biological findings from during the past decade expand on the known benefits of breastfeeding for women and children, whether they are rich or poor.
Collapse
Affiliation(s)
- Cesar G Victora
- International Center for Equity in Health, Post-Graduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, Brazil.
| | - Rajiv Bahl
- Department of Maternal, Newborn, Child and Adolescent Health (MCA), WHO, Geneva, Switzerland
| | - Aluísio J D Barros
- International Center for Equity in Health, Post-Graduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Giovanny V A França
- International Center for Equity in Health, Post-Graduate Programme in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Susan Horton
- Department of Economics, University of Waterloo, ON, Canada
| | - Julia Krasevec
- Data and Analytics Section, Division of Data, Research, and Policy, UNICEF, New York, NY, USA
| | - Simon Murch
- University Hospital Coventry and Warwickshire, Coventry, UK
| | - Mari Jeeva Sankar
- WHO Collaborating Centre for Training and Research in Newborn Care, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Neff Walker
- Institute for International Programs, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nigel C Rollins
- Department of Maternal, Newborn, Child and Adolescent Health (MCA), WHO, Geneva, Switzerland
| |
Collapse
|
93
|
Mode of Birth Influences Preterm Infant Intestinal Colonization With Bacteroides Over the Early Neonatal Period. Adv Neonatal Care 2015; 15:386-93. [PMID: 26551793 DOI: 10.1097/anc.0000000000000237] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Intestinal colonization during infancy is important to short- and long-term health outcomes. Bacteroides, an early member of the intestinal microbiome, is necessary for breaking down complex molecules within the intestine and function to assist the body's immune system in fighting against potentially harmful pathogens. Little is known about the colonization pattern of Bacteroides in preterm infants during the early neonatal period. PURPOSE This study measured Bacteroides colonization during the early neonatal period in a population of preterm infants, based on clinical factors including mode of birth, antibiotics, and nutrition. METHODS Bacterial DNA was isolated from 144 fecal samples from 29 preterm infants and analyzed using quantitative real-time polymerase chain reaction. Analyses included liner mixed models to determine which clinical factors affect Bacteroides colonization of the infant gut. RESULTS We found that infants born via vaginal canal had a higher rate of increase in Bacteroides than infants born via cesarean section (P < .001). We did not find significant associations between antibiotic administration and differences in nutritional exposures with Bacteroides colonization. IMPLICATIONS FOR PRACTICE These findings highlight the significant influence of mode of birth on Bacteroides colonization. While mode of birth is not always modifiable, these study findings may help develop interventions for preterm infants born via cesarean section aimed at overcoming delayed Bacteroides colonization. IMPLICATIONS FOR RESEARCH Greater study of the intestinal microbiome and the clinical factors relevant to the preterm infant is needed so that interventions may be developed and tested, resulting in optimal microbial and immune health.
Collapse
|
94
|
Vujkovic-Cvijin I, Swainson LA, Chu SN, Ortiz AM, Santee CA, Petriello A, Dunham RM, Fadrosh DW, Lin DL, Faruqi AA, Huang Y, Apetrei C, Pandrea I, Hecht FM, Pilcher CD, Klatt NR, Brenchley JM, Lynch SV, McCune JM. Gut-Resident Lactobacillus Abundance Associates with IDO1 Inhibition and Th17 Dynamics in SIV-Infected Macaques. Cell Rep 2015; 13:1589-97. [PMID: 26586432 DOI: 10.1016/j.celrep.2015.10.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/14/2015] [Accepted: 10/08/2015] [Indexed: 01/23/2023] Open
Abstract
Gut microbes can profoundly modulate mucosal barrier-promoting Th17 cells in mammals. A salient feature of HIV/simian immunodeficiency virus (SIV) immunopathogenesis is the loss of Th17 cells, which has been linked to increased activity of the immunomodulatory enzyme, indoleamine 2,3-dioxygenase 1 (IDO 1). The role of gut microbes in this system remains unknown, and the SIV-infected rhesus macaque provides a well-described model for HIV-associated Th17 loss and mucosal immune disruption. We observed a specific depletion of gut-resident Lactobacillus during acute and chronic SIV infection of rhesus macaques, which was also seen in early HIV-infected humans. This depletion in rhesus macaques correlated with increased IDO1 activity and Th17 loss. Macaques supplemented with a Lactobacillus-containing probiotic exhibited decreased IDO1 activity during chronic SIV infection. We propose that Lactobacillus species inhibit mammalian IDO1 and thus may help to preserve Th17 cells during pathogenic SIV infection, providing support for Lactobacillus species as modulators of mucosal immune homeostasis.
Collapse
Affiliation(s)
- Ivan Vujkovic-Cvijin
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Louise A Swainson
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Simon N Chu
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; University of California, Berkeley-University of California, San Francisco Joint Medical Program, San Francisco, CA 94143, USA
| | - Alexandra M Ortiz
- Laboratory of Molecular Microbiology, Program in Tissue Immunity and Repair and Immunopathogenesis Section, NIAID, NIH, Bethesda, MD 20892, USA
| | - Clark A Santee
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Annalise Petriello
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard M Dunham
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Douglas W Fadrosh
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Din L Lin
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ali A Faruqi
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yong Huang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cristian Apetrei
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ivona Pandrea
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Frederick M Hecht
- Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher D Pilcher
- Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nichole R Klatt
- Department of Pharmaceutics, University of Washington, Seattle, WA 98121, USA
| | - Jason M Brenchley
- Laboratory of Molecular Microbiology, Program in Tissue Immunity and Repair and Immunopathogenesis Section, NIAID, NIH, Bethesda, MD 20892, USA
| | - Susan V Lynch
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Joseph M McCune
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
95
|
|
96
|
Eco-Evo-Devo: developmental symbiosis and developmental plasticity as evolutionary agents. Nat Rev Genet 2015; 16:611-22. [PMID: 26370902 DOI: 10.1038/nrg3982] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The integration of research from developmental biology and ecology into evolutionary theory has given rise to a relatively new field, ecological evolutionary developmental biology (Eco-Evo-Devo). This field integrates and organizes concepts such as developmental symbiosis, developmental plasticity, genetic accommodation, extragenic inheritance and niche construction. This Review highlights the roles that developmental symbiosis and developmental plasticity have in evolution. Developmental symbiosis can generate particular organs, can produce selectable genetic variation for the entire animal, can provide mechanisms for reproductive isolation, and may have facilitated evolutionary transitions. Developmental plasticity is crucial for generating novel phenotypes, facilitating evolutionary transitions and altered ecosystem dynamics, and promoting adaptive variation through genetic accommodation and niche construction. In emphasizing such non-genomic mechanisms of selectable and heritable variation, Eco-Evo-Devo presents a new layer of evolutionary synthesis.
Collapse
|
97
|
Narayan NR, Méndez-Lagares G, Ardeshir A, Lu D, Van Rompay KKA, Hartigan-O'Connor DJ. Persistent effects of early infant diet and associated microbiota on the juvenile immune system. Gut Microbes 2015; 6:284-9. [PMID: 26177107 PMCID: PMC4615596 DOI: 10.1080/19490976.2015.1067743] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Early infant diet has significant impacts on the gut microbiota and developing immune system. We previously showed that breast-fed and formula-fed rhesus macaques develop significantly different gut microbial communities, which in turn are associated with different immune systems in infancy. Breast-fed animals manifested greater T cell activation and proliferation and harbored robust pools of T helper 17 (TH17) cells. These differences were sustained throughout the first year of life. Here we examine groups of juvenile macaques (approximately 3 to 5 y old), which were breast-fed or formula-fed in infancy. We demonstrate that juveniles breast-fed in infancy maintain immunologic differences into the fifth year of life, principally in CD8(+) memory T cell activation. Additionally, long-term correlation networks show that breast-fed animals maintain persistent relationships between immune subsets that are not seen in formula-fed animals. These findings demonstrate that infant feeding practices have continued influence on immunity for up to 3 to 5 y after birth and also reveal mechanisms for microbial modulation of the immune system.
Collapse
Affiliation(s)
- Nicole R Narayan
- California National Primate Research Center; University of California; Davis, CA USA,Department of Medical Microbiology and Immunology; University of California; Davis, CA USA
| | - Gema Méndez-Lagares
- California National Primate Research Center; University of California; Davis, CA USA,Department of Medical Microbiology and Immunology; University of California; Davis, CA USA
| | - Amir Ardeshir
- California National Primate Research Center; University of California; Davis, CA USA
| | - Ding Lu
- California National Primate Research Center; University of California; Davis, CA USA,Department of Medical Microbiology and Immunology; University of California; Davis, CA USA
| | - Koen K A Van Rompay
- California National Primate Research Center; University of California; Davis, CA USA
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center; University of California; Davis, CA USA,Department of Medical Microbiology and Immunology; University of California; Davis, CA USA,Division of Experimental Medicine; Department of Medicine; University of California; San Francisco, CA USA,Correspondence to: Dennis J Hartigan-O'Connor;
| |
Collapse
|
98
|
Selwyn FP, Cheng SL, Bammler TK, Prasad B, Vrana M, Klaassen C, Cui JY. Developmental Regulation of Drug-Processing Genes in Livers of Germ-Free Mice. Toxicol Sci 2015; 147:84-103. [PMID: 26032512 DOI: 10.1093/toxsci/kfv110] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Very little is known about the effect of gut microbiota on the ontogeny of drug-processing genes (DPGs) in liver. In this study, livers were harvested from conventional (CV) and germ-free (GF) male and female mice from 1 to 90 days of age. RNA-Seq in livers of 90-day-old male mice showed that xenobiotic metabolism was the most downregulated pathway within the mRNA transcriptome in absence of intestinal bacteria. In male livers, the mRNAs of 67 critical DPGs partitioned into 4 developmental patterns (real-time-quantitative polymerase chain reaction): Pattern-1 gradually increased to adult levels in livers of CV mice and were downregulated in livers of GF mice, as exemplified by the major drug-metabolizing enzymes cytochrome 3a (Cyp3a) family, which are prototypical pregnane X receptor (PXR)-target genes. Genes in Pattern-2 include Cyp1a2 (aryl hydrocarbon receptor-target gene), Cyp2c family, and Cyp2e1, which were all upregulated mainly at 90 days of age; as well as the peroxisome proliferator-activated receptor α (PPARα)-target genes Cyp4a family and Aldh3a2, which were upregulated not only in 90-days adult age, but also between neonatal and adolescent ages (from 1 to 30 days of age). Genes in Pattern-3 were enriched predominantly in livers of 15-day-old mice, among which the sterol-efflux transporter dimers Abcg5/Abcg8 were downregulated in GF mice. Genes in Pattern-4 were neonatal-enriched, among which the transporter Octn1 mRNA tended to be lower in GF mice at younger ages but higher in adult GF mice as compared with age-matched CV mice. Protein assays confirmed the downregulation of the PXR-target gene Cyp3a protein (Western-blot and liquid chromatography tandem mass spectroscopy), and decreased Cyp3a enzyme activities in male GF livers. Increased microsomal-Cyp4a proteins and nuclear-PPARα were also observed in male GF livers. Interestingly, in contrast to male livers, the mRNAs of Cyp2c or Cyp4a were not readily upregulated in female GF livers approaching adult age, suggesting the maturation of female-specific hormones interferes with the interactions between intestinal microbiota and DPG ontogeny. In conclusion, intestinal microbiota markedly impacts the ontogeny of many hepatic DPGs in a gender-specific manner.
Collapse
Affiliation(s)
| | | | - Theo K Bammler
- *Department of Environmental & Occupational Health Sciences and
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Marc Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195
| | - Curtis Klaassen
- *Department of Environmental & Occupational Health Sciences and
| | - Julia Yue Cui
- *Department of Environmental & Occupational Health Sciences and
| |
Collapse
|
99
|
Subramanian S, Blanton LV, Frese SA, Charbonneau M, Mills DA, Gordon JI. Cultivating healthy growth and nutrition through the gut microbiota. Cell 2015; 161:36-48. [PMID: 25815983 DOI: 10.1016/j.cell.2015.03.013] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Indexed: 12/21/2022]
Abstract
Microbiota assembly is perturbed in children with undernutrition, resulting in persistent microbiota immaturity that is not rescued by current nutritional interventions. Evidence is accumulating that this immaturity is causally related to the pathogenesis of undernutrition and its lingering sequelae. Preclinical models in which human gut communities are replicated in gnotobiotic mice have provided an opportunity to identify and predict the effects of different dietary ingredients on microbiota structure, expressed functions, and host biology. This capacity sets the stage for proof-of-concept tests designed to deliberately shape the developmental trajectory and configurations of microbiota in children representing different geographies, cultural traditions, and states of health. Developing these capabilities for microbial stewardship is timely given the global health burden of childhood undernutrition, the effects of changing eating practices brought about by globalization, and the realization that affordable nutritious foods need to be developed to enhance our capacity to cultivate healthier microbiota in populations at risk for poor nutrition.
Collapse
Affiliation(s)
- Sathish Subramanian
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Laura V Blanton
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Steven A Frese
- Departments of Food Science & Technology and Viticulture & Enology, University of California, Davis, Davis, CA 95616, USA
| | - Mark Charbonneau
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - David A Mills
- Departments of Food Science & Technology and Viticulture & Enology, University of California, Davis, Davis, CA 95616, USA
| | - Jeffrey I Gordon
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
100
|
Oxford KL, Dela Pena-Ponce MGA, Jensen K, Eberhardt MK, Spinner A, Van Rompay KK, Rigdon J, Mollan KR, Krishnan VV, Hudgens MG, Barry PA, De Paris K. The interplay between immune maturation, age, chronic viral infection and environment. IMMUNITY & AGEING 2015; 12:3. [PMID: 25991918 PMCID: PMC4436863 DOI: 10.1186/s12979-015-0030-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/24/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND The worldwide increase in life expectancy has been associated with an increase in age-related morbidities. The underlying mechanisms resulting in immunosenescence are only incompletely understood. Chronic viral infections, in particular infection with human cytomegalovirus (HCMV), have been suggested as a main driver in immunosenescence. Here, we propose that rhesus macaques could serve as a relevant model to define the impact of chronic viral infections on host immunity in the aging host. We evaluated whether chronic rhesus CMV (RhCMV) infection, similar to HCMV infection in humans, would modulate normal immunological changes in the aging individual by taking advantage of the unique resource of rhesus macaques that were bred and raised to be Specific Pathogen Free (SPF-2) for distinct viruses. RESULTS Our results demonstrate that normal age-related immunological changes in frequencies, activation, maturation, and function of peripheral blood cell lymphocytes in humans occur in a similar manner over the lifespan of rhesus macaques. The comparative analysis of age-matched SPF-2 and non-SPF macaques that were housed under identical conditions revealed distinct differences in certain immune parameters suggesting that chronic pathogen exposure modulated host immune responses. All non-SPF macaques were infected with RhCMV, suggesting that chronic RhCMV infection was a major contributor to altered immune function in non-SPF macaques, although a causative relationship was not established and outside the scope of these studies. Further, we showed that immunological differences between SPF-2 and non-SPF macaques were already apparent in adolescent macaques, potentially predisposing RhCMV-infected animals to age-related pathologies. CONCLUSIONS Our data validate rhesus macaques as a relevant animal model to study how chronic viral infections modulate host immunity and impact immunosenescence. Comparative studies in SPF-2 and non-SPF macaques could identify important mechanisms associated with inflammaging and thereby lead to new therapies promoting healthy aging in humans.
Collapse
Affiliation(s)
- Kristie L Oxford
- Center of Comparative Medicine, University of California, Davis, California USA
| | - Myra Grace A Dela Pena-Ponce
- Department of Microbiology and Immunology, University of North Carolina, Burnett-Womack Bldg, 160 Dental Circle, Chapel Hill, NC 27599-7292 USA
| | - Kara Jensen
- Department of Microbiology and Immunology, University of North Carolina, Burnett-Womack Bldg, 160 Dental Circle, Chapel Hill, NC 27599-7292 USA
| | - Meghan K Eberhardt
- Center of Comparative Medicine, University of California, Davis, California USA
| | - Abigail Spinner
- California National Primate Research Center, University of California, Davis, California USA
| | - Koen Ka Van Rompay
- California National Primate Research Center, University of California, Davis, California USA
| | - Joseph Rigdon
- Gillings School of Public Health, University of North Carolina, Chapel Hill, North Carolina USA
| | - Katie R Mollan
- Gillings School of Public Health, University of North Carolina, Chapel Hill, North Carolina USA.,Center for AIDS Research, University of North Carolina, Chapel Hill, North Carolina USA
| | - V V Krishnan
- Department of Pathology and Laboratory Medicine, University of California, Davis, California USA
| | - Michael G Hudgens
- Gillings School of Public Health, University of North Carolina, Chapel Hill, North Carolina USA.,Center for AIDS Research, University of North Carolina, Chapel Hill, North Carolina USA
| | - Peter A Barry
- Center of Comparative Medicine, University of California, Davis, California USA.,California National Primate Research Center, University of California, Davis, California USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Burnett-Womack Bldg, 160 Dental Circle, Chapel Hill, NC 27599-7292 USA.,Center for AIDS Research, University of North Carolina, Chapel Hill, North Carolina USA
| |
Collapse
|