1
|
Ohtsuka Y, Shimizu T. Immaturity of gut functions and induction of tolerance during early infancy. Pediatr Int 2025; 67:e15855. [PMID: 39739631 DOI: 10.1111/ped.15855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/02/2025]
Abstract
Tolerance to foreign molecules is primarily induced through three pathways: anergy, active suppression, and clonal deletion. The immaturity of gut functions, including digestion and barrier protection against foreign molecules during early infancy, is closely linked to the induction of tolerance. A significant number of undigested peptides can pass through leaky gut walls during this period, making it an opportune time to introduce active suppression and clonal deletion in the intestine. Increased production and expression of TGF-β and CXCL13 with lymphoid hyperplasia in early infancy can be a convenient way to introduce tolerance and immunoglobulin A (IgA) synthesis against food and other antigens to protect hosts from allergic diseases. In this article we will discuss the immaturity of gut functions and mucosal immunity in infancy and its relationship to the introduction of tolerance.
Collapse
Affiliation(s)
- Yoshikazu Ohtsuka
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Borghesi A. Life-threatening infections in human newborns: Reconciling age-specific vulnerability and interindividual variability. Cell Immunol 2024; 397-398:104807. [PMID: 38232634 DOI: 10.1016/j.cellimm.2024.104807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024]
Abstract
In humans, the interindividual variability of clinical outcome following exposure to a microorganism is immense, ranging from silent infection to life-threatening disease. Age-specific immune responses partially account for the high incidence of infection during the first 28 days of life and the related high mortality at population level. However, the occurrence of life-threatening disease in individual newborns remains unexplained. By contrast, inborn errors of immunity and their immune phenocopies are increasingly being discovered in children and adults with life-threatening viral, bacterial, mycobacterial and fungal infections. There is a need for convergence between the fields of neonatal immunology, with its in-depth population-wide characterization of newborn-specific immune responses, and clinical immunology, with its investigations of infections in patients at the cellular and molecular levels, to facilitate identification of the mechanisms of susceptibility to infection in individual newborns and the design of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, EU, Italy; School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| |
Collapse
|
3
|
Duess JW, Sampah ME, Lopez CM, Tsuboi K, Scheese DJ, Sodhi CP, Hackam DJ. Necrotizing enterocolitis, gut microbes, and sepsis. Gut Microbes 2023; 15:2221470. [PMID: 37312412 PMCID: PMC10269420 DOI: 10.1080/19490976.2023.2221470] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease in premature infants and the leading cause of death and disability from gastrointestinal disease in this vulnerable population. Although the pathophysiology of NEC remains incompletely understood, current thinking indicates that the disease develops in response to dietary and bacterial factors in the setting of a vulnerable host. As NEC progresses, intestinal perforation can result in serious infection with the development of overwhelming sepsis. In seeking to understand the mechanisms by which bacterial signaling on the intestinal epithelium can lead to NEC, we have shown that the gram-negative bacterial receptor toll-like receptor 4 is a critical regulator of NEC development, a finding that has been confirmed by many other groups. This review article provides recent findings on the interaction of microbial signaling, the immature immune system, intestinal ischemia, and systemic inflammation in the pathogenesis of NEC and the development of sepsis. We will also review promising therapeutic approaches that show efficacy in pre-clinical studies.
Collapse
Affiliation(s)
- Johannes W. Duess
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Maame E. Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Carla M. Lopez
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Koichi Tsuboi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Daniel J. Scheese
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Chhinder P. Sodhi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
4
|
Eddens T, Parks OB, Williams JV. Neonatal Immune Responses to Respiratory Viruses. Front Immunol 2022; 13:863149. [PMID: 35493465 PMCID: PMC9047724 DOI: 10.3389/fimmu.2022.863149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Respiratory tract infections are a leading cause of morbidity and mortality in newborns, infants, and young children. These early life infections present a formidable immunologic challenge with a number of possibly conflicting goals: simultaneously eliminate the acute pathogen, preserve the primary gas-exchange function of the lung parenchyma in a developing lung, and limit long-term sequelae of both the infection and the inflammatory response. The latter has been most well studied in the context of childhood asthma, where multiple epidemiologic studies have linked early life viral infection with subsequent bronchospasm. This review will focus on the clinical relevance of respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and rhinovirus (RV) and examine the protective and pathogenic host responses within the neonate.
Collapse
Affiliation(s)
- Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Division of Allergy/Immunology, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Olivia B. Parks
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - John V. Williams
- Division of Pediatric Infectious Diseases, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
5
|
Abstract
The neonatal period and early infancy are times of increased vulnerability to infection. The immune system of infants undergoes rapid changes and a number of factors can influence the maturation and function of the early infant immune system, amongst these factors are maternal infections and immunity. Infants who are HIV-exposed, but uninfected show important immune alterations, which are likely to be associated with the increased morbidity and mortality observed in these infants. Maternally derived antibodies are crucial in early life to protect infants from infection during the time when their own immune system is becoming more experienced and fully mature. However, maternal antibodies can also interfere with the infant's own antibody responses to primary vaccination. Preterm infants are particularly vulnerable to infection, having not had the opportunity to benefit from the transplacental transfer of maternal antibodies in late pregnancy. In addition, further differences have been observed in the innate and adaptive immune system between preterm and term infants. Here, we focus on maternal influences on the infant immune system, using HIV and maternal vaccination as examples and finish by considering how prematurity impacts infant immune responses to vaccination.
Collapse
Affiliation(s)
- Petra Zimmermann
- From the Department of Paediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Christine E Jones
- Faculty of Medicine and Institute for Life Sciences, University of Southampton and NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
6
|
Sampah MES, Hackam DJ. Prenatal Immunity and Influences on Necrotizing Enterocolitis and Associated Neonatal Disorders. Front Immunol 2021; 12:650709. [PMID: 33968047 PMCID: PMC8097145 DOI: 10.3389/fimmu.2021.650709] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.
Collapse
Affiliation(s)
| | - David J. Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine , Baltimore, MD, United States
| |
Collapse
|
7
|
Sampah MES, Hackam DJ. Dysregulated Mucosal Immunity and Associated Pathogeneses in Preterm Neonates. Front Immunol 2020; 11:899. [PMID: 32499778 PMCID: PMC7243348 DOI: 10.3389/fimmu.2020.00899] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Many functions of the immune system are impaired in neonates, allowing vulnerability to serious bacterial, viral and fungal infections which would otherwise not be pathogenic to mature individuals. This vulnerability is exacerbated in compromised newborns such as premature neonates and those who have undergone surgery or who require care in an intensive care unit. Higher susceptibility of preterm neonates to infections is associated with delayed immune system maturation, with deficiencies present in both the innate and adaptive immune components. Here, we review recent insights into early life immunity, and highlight features associated with compromised newborns, given the challenges of studying neonatal immunity in compromised neonates due to the transient nature of this period of life, and logistical and ethical obstacles posed by undertaking studies newborns and infants. Finally, we highlight how the unique immunological characteristics of the premature host play key roles in the pathogenesis of diseases that are unique to this population, including necrotizing enterocolitis and the associated sequalae of lung and brain injury.
Collapse
Affiliation(s)
- Maame Efua S Sampah
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David J Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Teixeira FME, Pietrobon AJ, Oliveira LDM, Oliveira LMDS, Sato MN. Maternal-Fetal Interplay in Zika Virus Infection and Adverse Perinatal Outcomes. Front Immunol 2020; 11:175. [PMID: 32117303 PMCID: PMC7033814 DOI: 10.3389/fimmu.2020.00175] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022] Open
Abstract
During pregnancy, the organization of complex tolerance mechanisms occurs to assure non-rejection of the semiallogeneic fetus. Pregnancy is a period of vulnerability to some viral infections, mainly during the first and second trimesters, that may cause congenital damage to the fetus. Recently, Zika virus (ZIKV) infection has gained great notoriety due to the occurrence of congenital ZIKV syndrome, characterized by fetal microcephaly, which results from the ability of ZIKV to infect placental cells and neural precursors in the fetus. Importantly, in addition to the congenital effects, studies have shown that perinatal ZIKV infection causes a number of disorders, including maculopapular rash, conjunctivitis, and arthralgia. In this paper, we contextualize the immunological aspects involved in the maternal-fetal interface and vulnerability to ZIKV infection, especially the alterations resulting in perinatal outcomes. This highlights the need to develop protective maternal vaccine strategies or interventions that are capable of preventing fetal or even neonatal infection.
Collapse
Affiliation(s)
- Franciane Mouradian Emidio Teixeira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Anna Julia Pietrobon
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luana de Mendonça Oliveira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luanda Mara da Silva Oliveira
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil
| | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, LIM-56, Department of Dermatology, School of Medicine and Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Dreschers S, Ohl K, Schulte N, Tenbrock K, Orlikowsky TW. Impaired functional capacity of polarised neonatal macrophages. Sci Rep 2020; 10:624. [PMID: 31953452 PMCID: PMC6968972 DOI: 10.1038/s41598-019-56928-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/17/2019] [Indexed: 01/08/2023] Open
Abstract
Neonatal sepsis is accompanied by impaired apoptotic depletion of monocytes and macrophages (MΦ), aberrant cytokine production, impaired cell metabolism, and sustained inflammation. Macrophage-colony stimulating factor (M-CSF) triggers the differentiation from monocytes into MΦ (MΦ-0). Interleukin-10 (IL10) and Interferon-gamma (IFNy) further differentiate MΦ subpopulations, the anti-inflammatory MΦ-IL10 and the pro-inflammatory MΦ-IFNy subtype. We previously have shown significant differences between adult (PBMΦ) and cord blood (CBMΦ) in the metabolism of all subtypes. To test the hypothesis whether the competence to differentiate monocytes into MΦ-0 and to polarise into MΦ-IFNy and MΦ-IL10 was diminished in CBMΦ as compared to PBMΦ, we polarised monocytes by cultivation with M-CSF for 72 h, followed by stimulation with IFNy or IL10, for 48 h. After flow cytometry based immunotyping, we tested four functions: Phagocytosis of GFP-E. coli, uptake of erythrocytes, T-cell proliferation, induction of regulatory T-cells as well as phosphorylation analysis of AKT and STAT1/STAT3. Phosphorylation of STAT-1 and STAT-3, obligatory to differentiate into MΦ-IFNγ, MΦ-0 and MΦ-IL10, was found to be aberrant in CBMΦ. Whereas infected MΦ-0 showed identical phagocytic indices and intracellular degradation, TLR4-expression, NFkB up-regulation, IL10-, IL6-, and TNFα production of CBMΦ-0 were reduced. In addition, the capacity to bind aged erythrocytes and the consecutive IL10 production was lower in CBMΦ-IL10. Polarised PBMΦ-IFNy and PBMΦ-IL10 expressed higher levels of co-stimulatory receptors (CD80, CD86), had a higher capacity to stimulate T-cells and induced higher amounts of regulatory T-cells (all p < 0.05 vs. corresponding CBMΦ). Hypoxia-inducible-factor-1α (HIF-1α) was stronger expressed in CBMΦ-IFNy and upregulated in infected CBMΦ-0, whereas heme-oxygenase 1 (HO-1) expression was similar to adult PBMΦ. Neonatal MΦ-0, MΦ-IFNy and MΦ-IL10 polarisation is impaired with respect to phenotype and functions tested which may contribute to sustained inflammation in neonatal sepsis.
Collapse
Affiliation(s)
- Stephan Dreschers
- Section of Neonatology, University Children's Hospital, Aachen, 52074, Germany
| | - Kim Ohl
- Department of Pediatrics, RWTH Aachen University, Aachen, Germany
| | - Nora Schulte
- Department of Pediatrics, RWTH Aachen University, Aachen, Germany
| | - Klaus Tenbrock
- Department of Pediatrics, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
10
|
Fike AJ, Kumova OK, Carey AJ. Dissecting the defects in the neonatal CD8 + T-cell response. J Leukoc Biol 2019; 106:1051-1061. [PMID: 31260598 DOI: 10.1002/jlb.5ru0319-105r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
The neonatal period presents a complex scenario where the threshold of reactivity toward colonizing microbiota, maternal antigens, autoantigens, and pathogens must be carefully moderated and balanced. CD8+ T cells are critical for the response against intracellular bacteria and viruses, but this immune compartment maintains altered function relative to adult counterparts because of the unique challenges which infants face. Here, we review our current understanding of the factors which may promote the attenuation and altered function of the neonatal CD8+ T-cell response and potential avenues for future study. Specifically, we have focused on the neonatal CD8+ T-cell ontogeny, memory formation, TCR structure and repertoire, TCR inhibitory receptors, and the clinical implications of altered neonatal CD8+ T-cell function. Special emphasis has been placed on examining the response of preterm neonates relative to term neonates and adults.
Collapse
Affiliation(s)
- Adam J Fike
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ogan K Kumova
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alison J Carey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Huang S, Rabah H, Ferret-Bernard S, Le Normand L, Gaucher F, Guerin S, Nogret I, Le Loir Y, Chen XD, Jan G, Boudry G, Jeantet R. Propionic fermentation by the probiotic Propionibacterium freudenreichii to functionalize whey. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
12
|
Verhoeven D. Influence of Immunological Maturity on Respiratory Syncytial Virus-Induced Morbidity in Young Children. Viral Immunol 2018; 32:76-83. [PMID: 30499759 DOI: 10.1089/vim.2018.0121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a very frequent viral respiratory pathogen of the young (<5 years old) with a significant portion of young toddlers having been infected before 2 years of age. Although we understand that some of the morbidity associated with RSV in neonates is due to immunological maturation that favors immunosuppression over antiviral innate and/or adaptive immune responses, the rapid development of the immune system right after birth suggests that each age group (newborn, early infant, older infant, toddler, and older) may respond to the virus in different ways. In this study, we summarize the morbidity associated with infection in young children in the context of immunological maturation of monocytes/macrophages and the ramifications for poor innate control of viral pathogenesis. We also summarize key mechanisms that contribute to the diminished antiviral innate immune responses of these young children.
Collapse
Affiliation(s)
- David Verhoeven
- Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames , Iowa
| |
Collapse
|
13
|
Schneider A, Weier M, Herderschee J, Perreau M, Calandra T, Roger T, Giannoni E. IRF5 Is a Key Regulator of Macrophage Response to Lipopolysaccharide in Newborns. Front Immunol 2018; 9:1597. [PMID: 30050534 PMCID: PMC6050365 DOI: 10.3389/fimmu.2018.01597] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022] Open
Abstract
Infections are a leading cause of mortality and morbidity in newborns. The high susceptibility of newborns to infection has been associated with a limited capacity to mount protective immune responses. Monocytes and macrophages are involved in the initiation, amplification, and termination of immune responses. Depending on cues received from their environment, monocytes differentiate into M1 or M2 macrophages with proinflammatory or anti-inflammatory and tissue repair properties, respectively. The purpose of this study was to characterize differences in monocyte to macrophage differentiation and polarization between newborns and adults. Monocytes from umbilical cord blood of healthy term newborns and from peripheral blood of adult healthy subjects were exposed to GM-CSF or M-CSF to induce M1 or M2 macrophages. Newborn monocytes differentiated into M1 and M2 macrophages with similar morphology and expression of differentiation/polarization markers as adult monocytes, with the exception of CD163 that was expressed at sevenfold higher levels in newborn compared to adult M1 macrophages. Upon TLR4 stimulation, newborn M1 macrophages produced threefold to sixfold lower levels of TNF than adult macrophages, while production of IL-1-β, IL-6, IL-8, IL-10, and IL-23 was at similar levels as in adults. Nuclear levels of IRF5, a transcription factor involved in M1 polarization, were markedly reduced in newborns, whereas the NF-κB and MAP kinase pathways were not altered. In line with a functional role for IRF5, adenoviral-mediated IRF5 overexpression in newborn M1 macrophages restored lipopolysaccharide-induced TNF production. Altogether, these data highlight a distinct immune response of newborn macrophages and identify IRF5 as a key regulator of macrophage TNF response in newborns.
Collapse
Affiliation(s)
- Anina Schneider
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Manuela Weier
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Jacobus Herderschee
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Eric Giannoni
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
14
|
Effect of Pasteurella multocida Soluble Antigen Stimulation on the In Vitro Response of Peripheral Blood Mononuclear Cells of Holstein Calves. ACTA VET-BEOGRAD 2018. [DOI: 10.2478/acve-2018-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The expressions of cytokines mRNA, including interleukin-4 (IL-4), interleukin- 17A (IL-17A) and interferon-gamma (IFN-γ), their master regulatory transcription factors, and signal transducers and activator of transcription (STAT) stimulated in vitro with Pasteurella (P.) multocida soluble antigen were examined in peripheral blood mononuclear cells (PBMC) from Holstein calves. The healthy Holstein calves were divided into three groups; 2 weeks old (2W Group, N=8), 6 weeks old (6W Group, N=8), and 10 weeks old (10W Group, N=8). PBMC were stimulated in vitro by soluble antigen of P. multocida. There were significantly lower expressions of IFN-γ, IL-4, and STAT-6 mRNA of PBMC stimulated with P. multocida soluble antigen in the 2W Group compared to that in the 10W Group. Expression of IL-17A and IFN-γ in PBMC stimulated with P. multocida soluble antigen were significantly higher compared with the PBMC without stimulation in the 6W groups. The results of the present study demonstrated that 2W old calves had decreased cytokine expression of PBMC when in vitro stimulated with P. multocida soluble antigen in vitro.
Collapse
|
15
|
Collins A, Weitkamp JH, Wynn JL. Why are preterm newborns at increased risk of infection? Arch Dis Child Fetal Neonatal Ed 2018; 103:F391-F394. [PMID: 29382648 PMCID: PMC6013388 DOI: 10.1136/archdischild-2017-313595] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
One in 10 newborns will be born before completion of 36 weeks' gestation (premature birth). Infection and sepsis in preterm infants remain a significant clinical problem that represents a substantial financial burden on the healthcare system. Many factors predispose premature infants for having the greatest risk of developing and succumbing to infection as compared with all other age groups across the age spectrum. It is clear that the immune system of preterm infants exhibits distinct, rather than simply deficient, function as compared with more mature and older humans and that the immune function in preterm infants contributes to infection risk. While no single review can cover all aspects of immune function in this population, we will discuss key aspects of preterm neonatal innate and adaptive immune function that place them at high risk for developing infections and sepsis, as well as sepsis-associated morbidity and mortality.
Collapse
Affiliation(s)
- Amélie Collins
- Department of Pediatrics, Division of Neonatology, Columbia University, New York City, New York
| | - Jörn-Hendrik Weitkamp
- Department of Pediatrics, Division of Neonatology, Vanderbilt University, Nashville, Tennessee
| | - James L. Wynn
- Department of Pediatrics, Division of Neonatology, University of Florida, Gainesville, Florida,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
16
|
Restori KH, Srinivasa BT, Ward BJ, Fixman ED. Neonatal Immunity, Respiratory Virus Infections, and the Development of Asthma. Front Immunol 2018; 9:1249. [PMID: 29915592 PMCID: PMC5994399 DOI: 10.3389/fimmu.2018.01249] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/18/2018] [Indexed: 12/27/2022] Open
Abstract
Infants are exposed to a wide range of potential pathogens in the first months of life. Although maternal antibodies acquired transplacentally protect full-term neonates from many systemic pathogens, infections at mucosal surfaces still occur with great frequency, causing significant morbidity and mortality. At least part of this elevated risk is attributable to the neonatal immune system that tends to favor T regulatory and Th2 type responses when microbes are first encountered. Early-life infection with respiratory viruses is of particular interest because such exposures can disrupt normal lung development and increase the risk of chronic respiratory conditions, such as asthma. The immunologic mechanisms that underlie neonatal host-virus interactions that contribute to the subsequent development of asthma have not yet been fully defined. The goals of this review are (1) to outline the differences between the neonatal and adult immune systems and (2) to present murine and human data that support the hypothesis that early-life interactions between the immune system and respiratory viruses can create a lung environment conducive to the development of asthma.
Collapse
Affiliation(s)
- Katherine H Restori
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Bharat T Srinivasa
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Elizabeth D Fixman
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
17
|
Han M, Hong JY, Jaipalli S, Rajput C, Lei J, Hinde JL, Chen Q, Hershenson NM, Bentley JK, Hershenson MB. IFN-γ Blocks Development of an Asthma Phenotype in Rhinovirus-Infected Baby Mice by Inhibiting Type 2 Innate Lymphoid Cells. Am J Respir Cell Mol Biol 2017; 56:242-251. [PMID: 27679954 DOI: 10.1165/rcmb.2016-0056oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Early-life wheezing-associated infections with rhinovirus (RV) have been associated with asthma development in children. We have shown that RV infection of 6-day-old mice induces mucous metaplasia and airways hyperresponsiveness, which is dependent on IL-13, IL-25, and type 2 innate lymphoid cells (ILC2s). Infection of immature mice fails to induce lung IFN-γ expression, in contrast to mature 8-week-old mice with a robust IFN-γ response, consistent with the notion that deficient IFN-γ production in immature mice permits RV-induced type 2 immune responses. We therefore examined the effects of intranasal IFN-γ administration on RV-induced ILC2 expansion and IL-13 expression in 6-day-old BALB/c and IL-13 reporter mice. Airway responses were assessed by histology, immunofluorescence microscopy, quantitative polymerase chain reaction, ELISA, and flow cytometry. Lung ILC2s were also treated with IFN-γ ex vivo. We found that, compared with untreated RV-infected immature mice, IFN-γ treatment attenuated RV-induced IL-13 and Muc5ac mRNA expression and mucous metaplasia. IFN-γ also reduced ILC2 expansion and the percentage of IL-13-secreting ILC2s. IFN-γ had no effect on the mRNA or protein expression of IL-25, IL-33, or thymic stromal lymphoprotein. Finally, IFN-γ treatment of sorted ILC2s reduced IL-5, IL-13, IL-17RB, ST2, and GATA-3 mRNA expression. We conclude that, in immature mice, IFN-γ inhibits ILC2 expansion and IL-13 expression in vivo and ex vivo, thereby attenuating RV-induced mucous metaplasia. These findings demonstrate the antagonistic function of IFN-γ on ILC2 expansion and gene expression, the absence of which may contribute to the development of an asthma-like phenotype after early-life RV infection.
Collapse
Affiliation(s)
- Mingyuan Han
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Jun Young Hong
- 2 Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Suraj Jaipalli
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Charu Rajput
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Jing Lei
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Joanna L Hinde
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Qiang Chen
- Departments of 1 Pediatrics and Communicable Diseases, and
| | | | | | - Marc B Hershenson
- Departments of 1 Pediatrics and Communicable Diseases, and.,2 Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
18
|
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
|
19
|
Jung J, Gleave Parson M, Kraft JD, Lyda L, Kobe B, Davis C, Robinson J, Peña MMO, Robinson CM. Elevated interleukin-27 levels in human neonatal macrophages regulate indoleamine dioxygenase in a STAT-1 and STAT-3-dependent manner. Immunology 2016; 149:35-47. [PMID: 27238498 PMCID: PMC4981608 DOI: 10.1111/imm.12625] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/24/2016] [Accepted: 05/27/2016] [Indexed: 12/12/2022] Open
Abstract
Microbial infections are a major cause of infant mortality as a result of limitations in immune defences. Interleukin-27 (IL-27) is a heterodimeric cytokine produced primarily by leucocytes and is immunosuppressive toward lymphocytes and leucocytes. Our laboratory demonstrated that human neonatal macrophages express IL-27 more abundantly than adult macrophages. Similarly in mice, IL-27 expression is elevated early in life and maintained through infancy. To determine IL-27-regulated mechanisms that may limit immunity, we evaluated the expression of a number of genes in response to this cytokine in primary human neonatal macrophages. Indoleamine 2,3-dioxygenase (IDO) gene expression was increased dose-responsively by IL-27. We have previously demonstrated inhibition of T-cell proliferation and cytokine production by neonatal macrophage-generated IL-27, and IDO is often implicated in this negative regulation. An increase in IDO protein was demonstrated by immunofluorescence microscopy and was consistent with increased enzyme activity following treatment with IL-27. Inclusion of a soluble receptor to neutralize endogenous IL-27, decreased IDO expression and activity compared with untreated macrophages. In response to IL-27, neonatal macrophages phosphorylate signal transdcuer and activator of transcription 1 (STAT-1) and STAT-3. Both transcription factors are recruited to the IDO regulatory region. STAT-3 dominates during steady-state regulation by lower levels of endogenous IL-27 production. A shift to enhanced STAT-1 recruitment occurs during increased levels of exogenously supplied IL-27. These data suggest an interesting interplay of STAT-1 and STAT-3 to regulate IDO activity and immunosuppression in response to different levels of IL-27 in the microenvironment of the immune response that may further our understanding of this interesting cytokine.
Collapse
Affiliation(s)
- Joo‐Yong Jung
- Department of BiologyBriar Cliff UniversitySioux CityIAUSA
| | - Madeline Gleave Parson
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Jennifer D. Kraft
- Department of Pathology Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Logan Lyda
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Brianna Kobe
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| | - Celestia Davis
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Jembber Robinson
- Department of Pathology Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Maria Marjorette O. Peña
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Cory M. Robinson
- Biomedical Sciences DepartmentWest Virginia School of Osteopathic MedicineLewisburgWVUSA
| |
Collapse
|
20
|
Nicholson GC, Holloway RA, Leaker BR, Kilty I, Salganik M, Tan L, Barnes PJ, Donnelly LE. A novel flow cytometric-based method to measure kinase inhibition in sputum from COPD subjects. BMJ Open Respir Res 2016; 3:e000140. [PMID: 27403320 PMCID: PMC4932304 DOI: 10.1136/bmjresp-2016-000140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/05/2022] Open
Abstract
Introduction Janus kinases (JAKs) regulate inflammatory gene expression through phosphorylation of signal transducer and activator of transcription (STAT) proteins. Expression of STAT proteins is increased in chronic obstructive pulmonary disease (COPD), and may be involved in driving chronic inflammation. Oral JAK inhibitors are effective as anti-inflammatory therapy but exhibit dose-limiting adverse effects. Development of inhaled compounds would be enhanced by robust biomarkers that directly reflect the anti-inflammatory and pharmacological activity in the lung. Methods A novel flow cytometry assay was developed to measure STAT1 phosphorylation in sputum inflammatory cells. The standard sputum processing method was refined to improve sputum cell viability. The flow cytometric assay was used to assess the reproducibility of the measurement of STAT1 phosphorylation and the in vitro activity of a pan JAK-inhibitor on three separate visits in patients with COPD. Results Upregulation of STAT1 phosphorylation was measured following in vitro IFNγ stimulation of sputum macrophages (stimulated/unstimulated ratio 1.57; p<0.00001). Upregulation was inhibited following in vitro preincubation with a pan JAK-inhibitor (inhibited+stimulated/unstimulated ratio 0.97). STAT1 phosphorylation activity could only be measured in macrophages. Conclusions Sputum from patients with COPD can be used to reproducibly measure phospho-STAT expression in sputum macrophages. The flow cytometry-based method can be used to evaluate kinase inhibitors in vitro and subsequently in ex vivo studies. The assay is particularly useful for the assessment of inhaled compounds where whole blood assays may not be relevant.
Collapse
Affiliation(s)
| | - R A Holloway
- Airways Disease Section , National Heart & Lung Institute, Imperial College , London , UK
| | - B R Leaker
- Respiratory Clinical Trials Ltd , London , UK
| | - I Kilty
- Pfizer , Cambridge, Massachusetts , USA
| | | | - L Tan
- Pfizer , Cambridge, Massachusetts , USA
| | - P J Barnes
- Airways Disease Section , National Heart & Lung Institute, Imperial College , London , UK
| | - L E Donnelly
- Airways Disease Section , National Heart & Lung Institute, Imperial College , London , UK
| |
Collapse
|
21
|
Rivera LE, Kraiselburd E, Meléndez LM. Cystatin B and HIV regulate the STAT-1 signaling circuit in HIV-infected and INF-β-treated human macrophages. J Neurovirol 2016; 22:666-673. [PMID: 27137788 DOI: 10.1007/s13365-016-0443-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 03/20/2016] [Accepted: 04/04/2016] [Indexed: 11/28/2022]
Abstract
Cystatin B is a cysteine protease inhibitor that induces HIV replication in monocyte-derived macrophages (MDM). This protein interacts with signal transducer and activator of transcription (STAT-1) factor and inhibits the interferon (IFN-β) response in Vero cells by preventing STAT-1 translocation to the nucleus. Cystatin B also decreases the levels of tyrosine-phosphorylated STAT-1 (STAT-1PY). However, the mechanisms of cystatin B regulation on STAT-1 phosphorylation in MDM are unknown. We hypothesized that cystatin B inhibits IFN-β antiviral responses and induces HIV replication in macrophage reservoirs through the inhibition of STAT-1 phosphorylation. Macrophages were transfected with cystatin B siRNA prior to interferon-β treatment or infected with HIV-ADA to determine the effect of cystatin B modulation in STAT-1 localization and activation using immunofluorescence and proximity ligation assays. Cystatin B decreased STAT-1PY and its transportation to the nucleus, while HIV infection retained unphosphorylated STAT (USTAT-1) in the nucleus avoiding its exit to the cytoplasm for eventual phosphorylation. In IFN-β-treated MDM, cystatin B inhibited the nuclear translocation of both, USTAT-1 and STAT-1PY. These results demonstrate that cystatin B interferes with the STAT-1 signaling and IFN-β-antiviral responses perpetuating HIV in macrophage reservoirs.
Collapse
Affiliation(s)
- L E Rivera
- Universidad del Este, Carolina, PR, 00984, USA
| | - E Kraiselburd
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, 00935, USA
| | - L M Meléndez
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, 00935, USA.
| |
Collapse
|
22
|
Kumar SKM, Bhat BV. Distinct mechanisms of the newborn innate immunity. Immunol Lett 2016; 173:42-54. [PMID: 26994839 DOI: 10.1016/j.imlet.2016.03.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/23/2022]
Abstract
The ontogeny of immunity during early life is of high importance as it shapes the immune system for the entire course of life. The microbiome and the environment contribute to the development of immunity in newborns. As immune responses in newborns are predominantly less experienced they are increasingly susceptible to infections. Though the immune cells in newborns are in 'naïve' state, they have been shown to mount adult-like responses in several circumstances. The innate immunity plays a vital role in providing protection during the neonatal period. Various stimulants have been shown to enhance the potential and functioning of the innate immune cells in newborns. They are biased against the production of pro-inflammatory cytokines and this makes them susceptible to wide variety of intracellular pathogens. The adaptive immunity requires prior antigenic experience which is very limited in newborns. This review discusses in detail the characteristics of innate immunity in newborns and the underlying developmental and functional mechanisms involved in the immune response. A better understanding of the immunological milieu in newborns could help the medical fraternity to find novel methods for prevention and treatment of infection in newborns.
Collapse
Affiliation(s)
- S Kingsley Manoj Kumar
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry 605006, India.
| | - B Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry 605006, India.
| |
Collapse
|
23
|
The role of inflammatory cytokines as key modulators of neurogenesis. Trends Neurosci 2015; 38:145-57. [PMID: 25579391 DOI: 10.1016/j.tins.2014.12.006] [Citation(s) in RCA: 254] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/20/2014] [Accepted: 12/08/2014] [Indexed: 12/20/2022]
Abstract
Neurogenesis is an important process in the regulation of brain function and behaviour, highly active in early development and continuing throughout life. Recent studies have shown that neurogenesis is modulated by inflammatory cytokines in response to an activated immune system. To disentangle the effects of the different cytokines on neurogenesis, here we summarise and discuss in vitro studies on individual cytokines. We show that inflammatory cytokines have both a positive and negative role on proliferation and neuronal differentiation. Hence, this strengthens the notion that inflammation is involved in molecular and cellular mechanisms associated with complex cognitive processes and, therefore, that alterations in brain-immune communication are relevant to the development of neuropsychiatric disorders.
Collapse
|
24
|
De Kleer I, Willems F, Lambrecht B, Goriely S. Ontogeny of myeloid cells. Front Immunol 2014; 5:423. [PMID: 25232355 PMCID: PMC4153297 DOI: 10.3389/fimmu.2014.00423] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/19/2014] [Indexed: 12/21/2022] Open
Abstract
Granulocytes, monocytes, macrophages, and dendritic cells (DCs) represent a subgroup of leukocytes, collectively called myeloid cells. During the embryonic development of mammalians, myelopoiesis occurs in a stepwise fashion that begins in the yolk sac and ends up in the bone marrow (BM). During this process, these early monocyte progenitors colonize various organs such as the brain, liver, skin, and lungs and differentiate into resident macrophages that will self-maintain throughout life. DCs are constantly replenished from BM precursors but can also arise from monocytes in inflammatory conditions. In this review, we summarize the different types of myeloid cells and discuss new insights into their early origin and development in mice and humans from fetal to adult life. We specifically focus on the function of monocytes, macrophages, and DCs at these different developmental stages and on the intrinsic and environmental influences that may drive these adaptations.
Collapse
Affiliation(s)
- Ismé De Kleer
- VIB Inflammation Research Center, University of Ghent , Ghent , Belgium ; Department of Respiratory Medicine, University Hospital Ghent , Ghent , Belgium ; Department of Pulmonary Medicine, Erasmus MC , Rotterdam , Netherlands
| | - Fabienne Willems
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Charleroi , Belgium
| | - Bart Lambrecht
- VIB Inflammation Research Center, University of Ghent , Ghent , Belgium ; Department of Respiratory Medicine, University Hospital Ghent , Ghent , Belgium
| | - Stanislas Goriely
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles , Charleroi , Belgium
| |
Collapse
|
25
|
Nupponen I, Kuuliala A, Siitonen S, Repo H, Kuuliala K. Cord Blood Monocytes, Neutrophils and Lymphocytes from Preterm and Full-Term Neonates Show Multiple Aberrations in Signalling Profiles Measured Using Phospho-Specific Whole-Blood Flow Cytometry. Scand J Immunol 2013; 78:426-38. [DOI: 10.1111/sji.12094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 06/25/2013] [Indexed: 11/29/2022]
Affiliation(s)
- I. Nupponen
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki Finland
- Department of Neonatology; Hospital for Children and Adolescents; Helsinki University Central Hospital; Helsinki Finland
| | - A. Kuuliala
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki Finland
| | - S. Siitonen
- Laboratory Services (HUSLAB); Helsinki University Central Hospital; Helsinki Finland
| | - H. Repo
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki Finland
| | - K. Kuuliala
- Department of Bacteriology and Immunology; Haartman Institute; University of Helsinki; Helsinki Finland
| |
Collapse
|
26
|
Walter J, Dihné M. Species-dependent differences of embryonic stem cell-derived neural stem cells after Interferon gamma treatment. Front Cell Neurosci 2012; 6:52. [PMID: 23162429 PMCID: PMC3492763 DOI: 10.3389/fncel.2012.00052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 10/17/2012] [Indexed: 12/23/2022] Open
Abstract
Pluripotent stem cell (pSC)-derived, neural stem cells (NSCs) are actually extensively explored in the field of neuroregeneration and to clarify disease mechanisms or model neurological diseases in vitro. Regarding the latter, proliferation and differentiation of pSC-derived NSCs are investigated under the influence of a variety of different substances among them key players of inflammation. However, results generated on a murine genetic background are not always representative for the human situation which increasingly leads to the application of human cell culture systems derived from human pSCs. We investigated here, if the recently described interferon gamma (IFNγ)-induced dysregulated neural phenotype characterized by simultaneous expression of glial and neuronal markers on murine NSCs (Walter et al., 2011, 2012) can also be found on a human genetic background. For this purpose, we performed experiments with human embryonic stem cell-derived NSCs. We could show that the IFNγ-induced dysregulated neural phenotype cannot be induced in human NSCs. This difference occurs, although typical genes like signal transducers and activators of transcription 1 (Stat 1) or interferon regulatory factor 9 (IRF-9) are similarly regulated by IFNγ in both, murine and human populations. These results illustrate that fundamental differences between murine and human neural populations exist in vitro, independent of anatomical system-related properties.
Collapse
Affiliation(s)
- Janine Walter
- Department of Neurology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany ; Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard-Karls-University Tübingen, Germany
| | | |
Collapse
|
27
|
Nyakeriga AM, Garg H, Joshi A. TCR-induced T cell activation leads to simultaneous phosphorylation at Y505 and Y394 of p56(lck) residues. Cytometry A 2012; 81:797-805. [PMID: 22674786 DOI: 10.1002/cyto.a.22070] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/12/2012] [Accepted: 04/23/2012] [Indexed: 11/11/2022]
Abstract
Biochemical studies have demonstrated that phosphorylation of lymphocyte cell kinase (p56(lck) ) is crucial for activation of signaling cascades following T cell receptor (TCR) stimulation. However, whether phosphorylation/dephosphorylation of the activating or inhibitory tyrosine residues occurs upon activation is controversial. Recent advances in intracellular staining of phospho-epitopes and cytometric analysis, requiring few cells, have opened up novel avenues for the field of immunological signaling. Here, we assessed p56(lck) phosphorylation, using a multiparameter flow-cytometric based detection method following T cell stimulation. Fixation and permeabilization in conjunction with zenon labeling technology and/or fluorescently labeled antibodies against total p56(lck) or cognate phospho-tyrosine (pY) residues or surface receptors were used for detection purposes. Our observations showed that activation of Jurkat or primary human T cells using H(2) O(2) or TCR-induced stimulation led to simultaneous phosphorylation of the activating tyrosine residue, Y394 and the inhibitory tyrosine residue, Y505 of p56(lck) . This was followed by downstream calcium flux and expression of T cell activation markers; CD69 and CD40 ligand (CD40L). However, the extent of measurable activation readouts depended on the optimal stimulatory conditions (temperature and/or stimuli combinations). Treatment of cells with a p56(lck) -specific inhibitor, PP2, abolished phosphorylation at either residue in a dose-dependent manner. Taken together, these observations show that TCR-induced stimulation of T cells led to simultaneous phosphorylation of p56(lck) residues. This implies that dephosphorylation of Y505 is not crucial for p56(lck) activity. Also, it is clear that cytometric analysis provides for a rapid, sensitive, and quantitative method to supplement biochemical studies on p56(lck) signaling pathways in T cells at single cell level.
Collapse
Affiliation(s)
- Alice M Nyakeriga
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, Texas 79905, USA.
| | | | | |
Collapse
|
28
|
Werneke SW, Schilte C, Rohatgi A, Monte KJ, Michault A, Arenzana-Seisdedos F, Vanlandingham DL, Higgs S, Fontanet A, Albert ML, Lenschow DJ. ISG15 is critical in the control of Chikungunya virus infection independent of UbE1L mediated conjugation. PLoS Pathog 2011; 7:e1002322. [PMID: 22028657 PMCID: PMC3197620 DOI: 10.1371/journal.ppat.1002322] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 09/02/2011] [Indexed: 01/13/2023] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus that has caused significant disease in the Indian Ocean region since 2005. During this outbreak, in addition to fever, rash and arthritis, severe cases of CHIKV infection have been observed in infants. Challenging the notion that the innate immune response in infants is immature or defective, we demonstrate that both human infants and neonatal mice generate a robust type I interferon (IFN) response during CHIKV infection that contributes to, but is insufficient for, the complete control of infection. To characterize the mechanism by which type I IFNs control CHIKV infection, we evaluated the role of ISG15 and defined it as a central player in the host response, as neonatal mice lacking ISG15 were profoundly susceptible to CHIKV infection. Surprisingly, UbE1L⁻/⁻ mice, which lack the ISG15 E1 enzyme and therefore are unable to form ISG15 conjugates, displayed no increase in lethality following CHIKV infection, thus pointing to a non-classical role for ISG15. No differences in viral loads were observed between wild-type (WT) and ISG15⁻/⁻ mice, however, a dramatic increase in proinflammatory cytokines and chemokines was observed in ISG15⁻/⁻ mice, suggesting that the innate immune response to CHIKV contributes to their lethality. This study provides new insight into the control of CHIKV infection, and establishes a new model for how ISG15 functions as an immunomodulatory molecule in the blunting of potentially pathologic levels of innate effector molecules during the host response to viral infection.
Collapse
Affiliation(s)
- Scott W. Werneke
- Department of Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Clementine Schilte
- Department of Immunology, Unité Immunobiologie des Cellules Dendritiques, Institut Pasteur, Paris, France
- INSERM U818, Paris, France
| | - Anjali Rohatgi
- Department of Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kristen J. Monte
- Department of Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alain Michault
- Laboratoire de Microbiologie, Groupe Hospitalier Sud Réunion, Saint-Pierre, France
| | | | - Dana L. Vanlandingham
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
| | - Arnaud Fontanet
- Epidemiology of Emerging Infectious Diseases, Institut Pasteur, Paris, France
| | - Matthew L. Albert
- Department of Immunology, Unité Immunobiologie des Cellules Dendritiques, Institut Pasteur, Paris, France
- INSERM U818, Paris, France
- * E-mail: (DJL); (MLA)
| | - Deborah J. Lenschow
- Department of Medicine, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (DJL); (MLA)
| |
Collapse
|
29
|
Méhes L, Taskó S, Székely A, Tóth Á, Ungvári E, Erdei I, Dombrádi Z, Szabó J, Maródi L. Phagocytosis and intracellular killing of heterogeneous vancomycin-intermediate Staphylococcus aureus strains. J Med Microbiol 2011; 61:198-203. [PMID: 21959204 DOI: 10.1099/jmm.0.029421-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Risk factors for invasive infections by heterogeneous vancomycin-intermediate Staphylococcus aureus (hVISA) may involve resistance to opsonophagocytosis and bacterial killing. hVISA strains typically have a thickened cell wall with altered peptidoglycan cross-linking. To determine whether hVISA may be endowed with an increased resistance to phagocytosis, this study assessed the characteristics of uptake and killing by granulocytes of three hVISA strains. All isolates were analysed by multilocus sequence typing and staphylococcal chromosome cassette mec typing. One of the strains belonged to the Hungarian meticillin-resistant S. aureus (MRSA) clone ST239-MRSA-III and the other two to the New York/Japan MRSA clone ST5-MRSA-II. In the presence of 10 % normal serum, the extent of phagocytosis and killing by blood granulocytes was equivalent for hVISA, MRSA and meticillin-sensitive S. aureus (MSSA) strains. Using granulocytes and serum from one patient who survived hVISA infection, the rate of phagocytosis and killing was also found to be comparable to that by control cells in the presence of 10 % serum. However, phagocytosis and killing of hVISA and MRSA (ATCC 25923) strains by normal granulocytes was markedly decreased in the presence of low concentrations (1 and 2.5 %) of serum from the patient who survived hVISA infection compared with that found with normal human serum. These data suggest that hVISA and MRSA isolates may be more resistant to opsonophagocytosis and bacterial killing than MSSA isolates, at least in some cases.
Collapse
Affiliation(s)
- Leonóra Méhes
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Szilvia Taskó
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Annamária Székely
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Ákos Tóth
- Department of Bacteriology, National Center for Epidemiology, Budapest, Hungary
| | - Erika Ungvári
- Department of Phage and Molecular Typing, National Center for Epidemiology, Budapest, Hungary
| | - Irén Erdei
- Department of Dermatology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Dombrádi
- Department of Medical Microbiology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Judit Szabó
- Department of Medical Microbiology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - László Maródi
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
30
|
Immunité innée du nouveau-né — Spécificités physiologiques et conséquences cliniques. MEDECINE INTENSIVE REANIMATION 2011. [DOI: 10.1007/s13546-010-0018-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
31
|
Jalkanen SE, Vakkila J, Kreutzman A, Nieminen JK, Porkka K, Mustjoki S. Poor cytokine-induced phosphorylation in chronic myeloid leukemia patients at diagnosis is effectively reversed by tyrosine kinase inhibitor therapy. Exp Hematol 2010; 39:102-113.e1. [PMID: 20869423 DOI: 10.1016/j.exphem.2010.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 09/14/2010] [Accepted: 09/15/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE In chronic myeloid leukemia (CML), uncontrolled tyrosine kinase activity of the BCR-ABL1 oncoprotein results in aberrant signaling pathways and increased cell proliferation. Acquired immune tolerance to leukemic antigens further enables tumor cell expansion. Tyrosine kinase inhibitor (TKI) therapy interferes with the immunoregulatory system by targeting off-target kinases both in malignant and nonmalignant cells. The aim of this study was to analyze the immune cell function by phosphoprotein profiling in CML patients. MATERIALS AND METHODS Blood samples from diagnostic phase and TKI-treated patients were analyzed by multicolor phosphoprotein flow cytometry enabling measurements at the single-cell level. Both unstimulated baseline activation status and cytokine-induced responses were evaluated. RESULTS In diagnostic-phase and imatinib-treated patients, the baseline phosphoprotein activation status was similar to healthy controls. In dasatinib-treated patients, basal phosphoprotein levels were slightly decreased; in particular, the signal transduction and activator of transcription protein 3 pathway was affected in both myeloid and lymphoid cells. The activation responses to various cytokines, granulocyte-macrophage colony-stimulating factor in particular were significantly suppressed in untreated CML patients. During imatinib and dasatinib therapy, the aberrantly suppressed phosphorylation responses were normalized. CONCLUSIONS Cytokine responses are hampered in untreated CML patients, which may have an effect on various immunological processes in vivo. Interestingly, during TKI treatment, phosphorylation responses were normal, suggesting that TKI treatment does not alter the reactivity of healthy immune effector cells. However, dasatinib treatment was associated with diminished basal activation of the immunosuppressive signal transduction and activator of transcription protein 3 signaling pathway, which could have clinical significance in reversing the lymphocyte anergy against tumor cells.
Collapse
Affiliation(s)
- Sari E Jalkanen
- Hematology Research Unit, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
32
|
Melvan JN, Bagby GJ, Welsh DA, Nelson S, Zhang P. Neonatal sepsis and neutrophil insufficiencies. Int Rev Immunol 2010; 29:315-48. [PMID: 20521927 DOI: 10.3109/08830181003792803] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sepsis has continuously been a leading cause of neonatal morbidity and mortality despite current advances in chemotherapy and patient intensive care facilities. Neonates are at high risk for developing bacterial infections due to quantitative and qualitative insufficiencies of innate immunity, particularly granulocyte lineage development and response to infection. Although antibiotics remain the mainstay of treatment, adjuvant therapies enhancing immune function have shown promise in treating sepsis in neonates. This article reviews current strategies for the clinical management of neonatal sepsis and analyzes mechanisms underlying insufficiencies of neutrophil defense in neonates with emphasis on new directions for adjuvant therapy development.
Collapse
Affiliation(s)
- John Nicholas Melvan
- Department of Physiology and Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
33
|
Rowland RRR. The interaction between PRRSV and the late gestation pig fetus. Virus Res 2010; 154:114-22. [PMID: 20832434 PMCID: PMC7172144 DOI: 10.1016/j.virusres.2010.09.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 06/17/2010] [Indexed: 12/12/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) crosses the placenta during late gestation and productively infects the fetus. Virus replication and cytokine responses were measured in tissues of fetuses recovered at 109–112 days of gestation, just prior to parturition. At the time of recovery, gross anatomical abnormalities were evident in both infected and non-infected fetuses from the infected dams. Virus isolation and immunohistochemistry identified the thymus as the primary site of virus replication. Steady state RT-PCR amplification of inflammatory, Th1 and Th2 cytokines, showed elevated IFN-γ and TNF-α mRNAs in tissues from infected fetuses, which corresponded to elevated cytokine proteins in serum but not amniotic fluid. Further evidence for induction of immunity was found in the hyperplastic response of lymph nodes, which included the development of germinal centers occupied CDw75+ B cells. Collectively, these data support the notion that the immunocompetent fetus is capable of initiating an antiviral response, which is compartmentalized within the infected fetus. Furthermore, fetal pathology may not be a direct result of virus replication in the fetus.
Collapse
Affiliation(s)
- Raymond R R Rowland
- Department of Diagnostic Medicine and Pathobiology, 1800 Denison Ave, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
34
|
Wu S, Jin L, Vence L, Radvanyi LG. Development and application of 'phosphoflow' as a tool for immunomonitoring. Expert Rev Vaccines 2010; 9:631-43. [PMID: 20518718 DOI: 10.1586/erv.10.59] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Flow cytometry has revolutionized our ability to monitor immune responses by allowing us to simultaneously track a variety of cell surface and intracellular markers in discrete cell subsets in a highly sensitive and reproducible manner. This is especially critical in this new era of vaccinology trying to tackle the growing problems of chronic viral infections and cancer that not only evade host immune responses, but can negatively manipulate vaccine-induced immune responses. Thus, understanding how lymphocyte signaling is altered under normal and pathological conditions has become more critical. Over the last decade, a new flow cytometry technology called 'phosphoflow' (also sometimes called 'phosflow'), is rapidly developing for tracking multiple intracellular signaling molecules in the immune system at a single-cell level. Antibodies and reagents for tracking both tyrosine-phosphorylated and serine/threonine-phosphorylated signaling intermediaries in key immune signaling pathways have been developed, and phosphoflow is now starting to be applied to a wide variety of both preclinical and clinical studies on lymphocyte responses, as well as the functioning of cancer cells and virally infected cells. Here, we review the development of phosphoflow technology, its modern applications in the field of immunomonitoring and its current limitations. We then provide a perspective on the future of phosphoflow and a vision of how it can be applied to emerging critical questions in human vaccinology and public health.
Collapse
Affiliation(s)
- Sheng Wu
- Department of Melanoma Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
35
|
Pérez A, Bellón JM, Gurbindo MD, Muñoz-Fernández MÁ. Impairment of stimulation ability of very-preterm neonatal monocytes in response to lipopolysaccharide. Hum Immunol 2010; 71:151-7. [DOI: 10.1016/j.humimm.2009.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 11/11/2009] [Accepted: 11/11/2009] [Indexed: 12/24/2022]
|
36
|
Merkerova M, Vasikova A, Bruchova H, Libalova H, Topinka J, Balascak I, Sram RJ, Brdicka R. Differential gene expression in umbilical cord blood and maternal peripheral blood. Eur J Haematol 2009; 83:183-90. [PMID: 19500137 DOI: 10.1111/j.1600-0609.2009.01281.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Umbilical cord blood (UCB) has become a useful alternative source of hematopoietic stem cells for clinical and research applications. UCB represents neonatal blood and differs from adult blood in many aspects, displaying different cell composition and various features of cellular immaturity. To understand molecular basis of phenotypic differences between neonatal and adult blood, we studied variations in transcriptome of UCB and maternal peripheral blood (PB). METHODS Using Illumina microarrays, we determined gene expression profiles of UCB and PB samples obtained from 30 mothers giving birth to living baby. RESULTS Out of 20,589 tested genes, 424 genes were down-regulated and 417 genes were up-regulated in UCB compared with PB. Reduced expression of many immunity-related pathways (e.g. TLR pathway, Jak-STAT pathway, cytokine-cytokine receptor interaction) in neonatal blood cells may contribute to the poor response to antigens, increasing susceptibility to infections at the time of disappearance of protective maternal antibodies. On the other hand, overexpression of erythropoiesis-related genes (glycophorins, fetal hemoglobins, enzymes catalysing heme synthesis and erythrocyte differentiation) in UCB probably enforces red cell production in newborns. CONCLUSIONS Our study demonstrates that neonatal and maternal bloods show specific gene expression profiles, likely reflecting differences in phenotypes of immunologically immature and fully evolved hematopoietic cells.
Collapse
Affiliation(s)
- Michaela Merkerova
- Institute of Hematology and Blood Transfusion, 128 20 Prague 2, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
37
|
M'Rabet L, Vos AP, Boehm G, Garssen J. Breast-feeding and its role in early development of the immune system in infants: consequences for health later in life. J Nutr 2008; 138:1782S-1790S. [PMID: 18716187 DOI: 10.1093/jn/138.9.1782s] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Laura M'Rabet
- Utrecht Institute of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, 3508 TC Utrecht, The Netherlands
| | | | | | | |
Collapse
|
38
|
Tatad AMF, Nesin M, Peoples J, Cheung S, Lin H, Sison C, Perlman J, Cunningham-Rundles S. Cytokine expression in response to bacterial antigens in preterm and term infant cord blood monocytes. Neonatology 2008; 94:8-15. [PMID: 18097152 DOI: 10.1159/000112541] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Accepted: 08/20/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND Neonatal susceptibility to bacterial infection is associated with an immature immune system, but the role of different bacterial antigens in specific responses is largely unknown. OBJECTIVE To evaluate differences in intracellular cytokine response to physiologically relevant bacterial antigens in term and preterm infants as compared with adults. METHODS Cord blood samples from preterm and term neonates and adult peripheral blood samples were cultured ex vivo with and without whole heat-killed bacteria. Intracellular leukocyte production of interleukin (IL)-6, IL-10, IL-12, and IL-8 responses was assessed by flow cytometry. RESULTS Monocytes were the primary producers of all mediators. Escherichia coli was the most potent stimulant. Lactobacillus plantarum 299v activated fewer monocytes as compared with E. coli for all responses (p < 0.05), except for IL-12 in term neonates. IL-6 response to Staphylococcus epidermidis was lower in both groups of neonates as compared with adults (p = 0.023 and p = 0.001). IL-8 response to S. epidermidis was lower in term as compared with preterm neonates and adults (p = 0.003). IL-10 response to group B streptococci was lower in term neonates as compared with adults and higher in preterm as compared with term neonates (p = 0.015). CONCLUSIONS Monocytes from term neonates compared to preterm neonates show a downregulated anti-inflammatory response to specific bacteria. High neonatal response to pathogenic E. coli in the preterm infant could cause uncontrolled inflammatory response, while lower IL-6 response to S. epidermidis in neonates may indicate a basis for vulnerability to S. epidermidis infection.
Collapse
Affiliation(s)
- A M Francesca Tatad
- Department of Pediatrics, Host Defenses Program, Weill Cornell Medical College, Manhasset, NY 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Vakkila J, Nieminen U, Siitonen S, Turunen U, Halme L, Nuutinen H, Mustonen H, Puolakkainen P, Färkkilä M, Repo H. A novel modification of a flow cytometric assay of phosphorylated STAT1 in whole blood monocytes for immunomonitoring of patients on IFN alpha regimen. Scand J Immunol 2007; 67:95-102. [PMID: 18028288 DOI: 10.1111/j.1365-3083.2007.02028.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We explored whether episodes stimulating leucocytes in vivo could be tracked from whole blood samples by monitoring activation of STAT1 by flow cytometry. The method was tested in hepatitis C patients (n = 9) that were on interferon (IFN)alpha regimen. CD14+ monocytes responded strongly to IFNalpha/gamma being sensitive indicators for recent immune activation. At 45 min after s.c. IFNalpha 91% of monocytes were phosphorylated STAT1+. The frequency of responding cells decreased to a base level within 6 h. Monocytes, however, had a long-term deficient phosphorylated STAT1 response to IFNalphain vitro that in patients on standard IFNalpha regimen lasted for 48 h. In patients on pegylated IFNalpha the phosphorylated STAT1 response was completely absent. We conclude that whole blood analysis of STAT1 activation by flow cytometry is applicable to monitor immune cells in patient material.
Collapse
Affiliation(s)
- J Vakkila
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Weinberger B, Vetrano AM, Syed K, Murthy S, Hanna N, Laskin JD, Laskin DL. Influence of labor on neonatal neutrophil apoptosis, and inflammatory activity. Pediatr Res 2007; 61:572-7. [PMID: 17413861 DOI: 10.1203/pdr.0b013e318045be38] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Neutrophil apoptosis is impaired in neonates, and this contributes to prolonged inflammation and tissue injury in infants after infection or trauma. In the present studies, we investigated whether labor generates mediators that further suppress apoptosis. We found that neutrophil apoptosis was reduced in neonates exposed to labor, when compared with infants delivered by cesarean section before labor. This was not due to alterations in caspase-3 or inhibitor of apoptosis protein-2 (IAP-2). In contrast, labor primed neutrophils to express tumor necrosis factor alpha (TNF-alpha), suggesting that proinflammatory mediators contribute to reduced apoptosis after labor. Eicosanoids generated via cyclooxygenase-2 (Cox-2) and lipoxygenase (Lox) also regulate neutrophil apoptosis. 15-Lox, which generates proapoptotic lipoxins, but not Cox-2, was greater in neutrophils before labor, relative to cells exposed to labor. Anti-inflammatory eicosanoids exert their effects in part via peroxisome proliferator-activated receptor gamma (PPAR-gamma). Expression of gelatinase-associated lipocalin and catalase, two markers of PPAR-gamma activity, were increased in neonatal neutrophils before labor, relative to cells exposed to labor. These findings suggest that the anti-inflammatory environment is maintained before labor, in part, by eicosanoids. Although increased neutrophil longevity after labor is important for host defense in the immediate newborn period, it may contribute to inflammatory or oxidative injury in susceptible infants.
Collapse
Affiliation(s)
- Barry Weinberger
- Department of Pediatrics/Neonatology, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey 08903, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Velilla PA, Rugeles MT, Chougnet CA. Defective antigen-presenting cell function in human neonates. Clin Immunol 2006; 121:251-9. [PMID: 17010668 PMCID: PMC1764492 DOI: 10.1016/j.clim.2006.08.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 08/02/2006] [Accepted: 08/21/2006] [Indexed: 01/07/2023]
Abstract
Immaturity of the immune system has been suggested as an underlying factor for the high rate of morbidity and mortality from infections in newborns. Functional impairment of neonatal T cells is frequently quoted as the main underlying mechanism for such immaturity. However, recent studies suggest that neonatal antigen-presenting cells (APCs) also exhibit functional alterations, which could lead to secondary defects of adaptive T-cell responses. In this review, we summarize what is known on the functionality of APC at birth and during early childhood. Compared to adults, neonatal APCs display markers of immaturity and produce low levels of cytokines. Multiple factors could be involved in neonatal APC alteration, such as intrinsic immaturity, defective interaction between APCs and T cells and regulatory T-cell-mediated inhibition. Characterization of the relative contribution of each mechanism is clearly needed to better understand the functional capability of the neonatal immune system.
Collapse
Affiliation(s)
- Paula A Velilla
- Group Immunovirology, Biogenesis Corporation, University of Antioquia, Medellín, A.A. 1226 Colombia
| | | | | |
Collapse
|
42
|
Lanuti P, Marchisio M, Cantilena S, Paludi M, Bascelli A, Gaspari AR, Grifone G, Centurione MA, Papa S, Di Pietro R, Cataldi A, Miscia S, Bertagnolo V. A flow cytometry procedure for simultaneous characterization of cell DNA content and expression of intracellular protein kinase C-zeta. J Immunol Methods 2006; 315:37-48. [PMID: 16945385 DOI: 10.1016/j.jim.2006.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 06/15/2006] [Accepted: 06/26/2006] [Indexed: 02/05/2023]
Abstract
A selective involvement of protein kinase C-zeta (PKC-zeta) in the events regulating cell proliferation has been recently proposed. Here we report a flow cytometric method allowing the simultaneous association of intracellular PKC-zeta expression or phosphorylation with each cell cycle phase. Current methods for flow cytometry analysis were applied to several cell lines and compared to the method developed in our laboratory. The latter includes 2% paraformaldehyde (PFA), as fixing agent, a permeabilization/saturation step by means of a solution containing 150 mM NaCl, 5 mM EDTA, 50 mM Tris-HCl pH 7.4, 0.05% NP-40, 0.25% lambda-carrageenan and 0.02% NaN3, followed by labelling with a primary antibody (PKC-zeta or P-PKC-zeta) and with the appropriate FITC-conjugated secondary antibody. Cells processed by such a method disclosed no substantial modification of light scattering features with respect to live cells. In addition, stainability with anti-PKC-zeta or anti-P-PKC-zeta antibodies was well preserved while stoichiometric staining of DNA with PI enabled accurate cell cycle analysis. Results show that a distinct up-regulation of P-PKC-zeta in G2/M phase occurs. The method here described, therefore, represents a simple, reproducible and conservative assay for a simultaneous assessment of intracellular PKC or P-PKC modulations within each cell cycle phase.
Collapse
Affiliation(s)
- Paola Lanuti
- Cell Signalling Unit at the Department of Biomorphology, University G. d'Annunzio Chieti-Pescara, Chieti, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- László Maródi
- Department of Infectious and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Nagyerdei Krt. 98, H-4012 Debrecen, Hungary.
| |
Collapse
|
44
|
Maródi L. Innate cellular immune responses in newborns. Clin Immunol 2005; 118:137-44. [PMID: 16377252 DOI: 10.1016/j.clim.2005.10.012] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 09/28/2005] [Accepted: 10/03/2005] [Indexed: 02/06/2023]
Abstract
Innate immunity assures the first line of defense against pathogenic microorganisms. Innate immune responses induced by bacteria, fungi, or viral replication are triggered by granulocytes, monocytes, macrophages, dentritic cells, and natural killer cells. Neonatal deficiency of innate cellular immunity includes a decreased production of interferons, IL-12/IL-23, and IL-18, and other proinflammatory cytokines, an impaired type-1 response of macrophages to IFN-gamma, the most potent macrophage-activating agent in vivo, and to lipopolysaccharide, the primary constituent of the outer membrane of Gram-negative bacteria. An increasing body of evidence suggests impaired responses of neonatal monocytes and macrophages to multiple TLR ligands. This review will discuss recent advances in understanding innate cellular immunity in human neonates, with respect to selected aspects of immune functions that may be related to increased susceptibility to infections. Components of TLR signaling and the immune consequence that may result from neonatal deficiencies will be highlighted. A better understanding of innate immunity can make the development of techniques possible by which physicians more accurately tailor prevention and treatment of neonatal infections.
Collapse
Affiliation(s)
- László Maródi
- Department of Infectiology and Pediatric Immunology, Medical and Health Science Center, University of Debrecen, Nagyerdei Krt. 98, H-4012 Debrecen, Hungary.
| |
Collapse
|
45
|
Maecker H, Varfolomeev E, Kischkel F, Lawrence D, LeBlanc H, Lee W, Hurst S, Danilenko D, Li J, Filvaroff E, Yang B, Daniel D, Ashkenazi A. TWEAK Attenuates the Transition from Innate to Adaptive Immunity. Cell 2005; 123:931-44. [PMID: 16325585 DOI: 10.1016/j.cell.2005.09.022] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 08/02/2005] [Accepted: 09/08/2005] [Indexed: 12/11/2022]
Abstract
Innate immunity is the first line of defense against infection, protecting the host during the development of adaptive immunity and critically affecting the nature of the adaptive response. We show that, in contrast to tumor necrosis factor alpha (TNF-alpha), the related protein TWEAK attenuates the transition from innate to adaptive mechanisms. TWEAK-/- mice had overabundant natural killer (NK) cells and displayed hypersensitivity to bacterial endotoxin, with their innate immune cells producing excess interferon (IFN)-gamma and interleukin (IL)-12. TWEAK inhibited stimulation of the transcriptional activator STAT-1 and induced p65 nuclear factor (NF)-kappaB association with histone deacetylase 1, repressing cytokine production. TWEAK-/- mice developed oversized spleens with expanded memory and T helper 1 (TH1) subtype cells upon aging and mounted stronger innate and adaptive TH1-based responses against tumor challenge. Thus, TWEAK suppresses production of IFN-gamma and IL-12, curtailing the innate response and its transition to adaptive TH1 immunity.
Collapse
Affiliation(s)
- Heather Maecker
- Department of Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Krutzik PO, Clutter MR, Nolan GP. Coordinate analysis of murine immune cell surface markers and intracellular phosphoproteins by flow cytometry. THE JOURNAL OF IMMUNOLOGY 2005; 175:2357-65. [PMID: 16081806 DOI: 10.4049/jimmunol.175.4.2357] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recently, phosphospecific flow cytometry has emerged as a powerful tool to analyze intracellular signaling events in complex populations of cells because of its ability to simultaneously discriminate cell types based on surface marker expression and measure levels of intracellular phosphoproteins. This has provided novel insights into the cell- and pathway-specific nature of immune signaling. However, we and others have found that the fixation and permeabilization steps necessary for phosphoprotein analysis often negatively affect the resolution of cell types based on surface marker analysis and light scatter characteristics. Therefore, we performed a comprehensive profile of >35 different murine surface marker Abs to understand the effects of fixation and permeabilization on surface Ag staining. Fortuitously, approximately 80% of the Abs tested resolved cell populations of interest, although with decreased separation between positive and negative populations and at very different titers than those used on live cells. The other 20% showed either complete loss of separation between populations or loss of intermediately staining populations. We were able to rescue staining of several of these Ags by performing staining after fixation, but before permeabilization, although with limited fluorophore choices. Scatter characteristics of lymphocytes were well retained, but changed dramatically for monocyte and neutrophil populations. These results compile a comprehensive resource for researchers interested in applying phosphospecific flow cytometry to complex populations of cells while outlining steps necessary to successfully apply new surface marker Abs to this platform.
Collapse
Affiliation(s)
- Peter O Krutzik
- Department of Microbiology and Immunology, Baxter Laboratory of Genetic Pharmacology, Stanford University, Stanford, CA 94305, USA
| | | | | |
Collapse
|
47
|
Cantó E, Rodríguez-Sánchez JL, Vidal S. Naive CD4+ cells from cord blood can generate competent Th effector cells. Transplantation 2005; 80:850-858. [PMID: 16210975 DOI: 10.1097/01.tp.0000174135.32068.65] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Umbilical cord blood (UCB) cells have been increasingly used as a source of hematopoietic stem cells for allogeneic transplantation. Previous reports suggest that the low risk of graft-versus-host disease in patients that received cord blood cells seems related to the distinctive nature of cord blood T cells. METHODS To analyze the maturation of CD4+CD45RA+ cord blood cells, we performed an in vitro differentiation assay to compare the generation of Th effector cells strictly from UCB and adult peripheral blood (APB) CD4+CD45RA+ cells. RESULTS During the maturation into effector cells, UCB and APB cells acquired a comparable activation level determined by the expression pattern of CD69, CD40L, OX40 and CD62L as well as PD1 and CTLA-4 molecules. Moreover, the expression of CD45RO isoform was induced in most activated effector cells from both UCB and APB. OKT3-restimulated effector cells generated from naive UCB expressed higher levels of CD25 coinciding with the secretion of higher amounts of IL-2. Effector cells from both origins consisted of heterogeneous populations with similar frequencies of Th1 and Th2 cytokine producing cells, secreting equivalent levels of IL-4, IL-5 and IFNgamma. Although, higher levels of IL-10 were detected in the cytokine mRNA profile and in the supernatants of OKT3-restimulated UCB effector cells, blocking endogenous IL-10 with anti-IL-10 mAbs enhanced significantly the proliferative response of UCB as well as APB effector cells (P < 0.05). CONCLUSIONS These results indicated that Th effector cells generated from naive UCB cells were intrinsically as competent as naive APB to respond to TCR-mediated stimulation. In addition, UCB effector cells produced higher IL-10 but its inhibitory effect on proliferation may be partially compensated by the higher production of IL-2 and enhanced expression of CD25.
Collapse
Affiliation(s)
- Elisabet Cantó
- Department of Immunology, Institut Recerca i Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | | | | |
Collapse
|
48
|
Bladon J, Taylor P. Extracorporeal Photopheresis Differentially Regulates the Expression of Phosphorylated STAT-1 and STAT-5 in Treated Monocytes and T cells, Respectively. J Cutan Med Surg 2004; 8:148-56. [PMID: 15578129 DOI: 10.1007/s10227-004-0102-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Extracorporeal photopheresis (ECP) is effective in conditions with opposing immune etiology. ECP induces an immunomodulatory response through simultaneous monocyte activation and apoptosis induction of lymphocytes. However, ECP is also immunosuppressive, downregulating proinflammatory cytokines. Signal transducers and activators of transcription (STATs) are important mediators of cell-signaling systems, including cytokines. Ultraviolet (UV) immunosuppression is linked to reductions in cytokine-induced STAT phosphorylation. OBJECTIVE The aim of this study was to find whether ECP downregulates STAT phosphorylation. METHODS Pre- and post-ECP mononuclear cells from cutaneous T-cell lymphoma and chronic graft-versus-host disease patients were stimulated with either IFNbeta, IL2, or IL15. The percentage of IFNbeta-stimulated monocytes positive for phosphorylated STAT-1 (pSTAT-1) and the number of IL2- and IL15-stimulated T cells expressing phosphorylated STAT-5 (pSTAT-5) were determined at 0 and 24 h. RESULTS Post-ECP, pSTAT-1 levels in monocytes remained unchanged; however, at 24 h post-ECP the number of T cells expressing pSTAT-5 was reduced. CONCLUSION Following ECP, monocytes retain their ability to phosphorylate STAT-1, while pSTAT-5 expression is lost in lymphocytes. This differential effect of ECP may account for the diverse population of diseases that benefit.
Collapse
Affiliation(s)
- John Bladon
- Department of Haematology, Rotherham General Hospital, S60 2UD, South Yorkshire, United Kingdom.
| | | |
Collapse
|
49
|
Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol 2004; 4:553-64. [PMID: 15229474 DOI: 10.1038/nri1394] [Citation(s) in RCA: 752] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Becky Adkins
- Department of Microbiology and Immunology, University of Miami Medical School, Miami, Florida 33136, USA.
| | | | | |
Collapse
|
50
|
Krutzik PO, Irish JM, Nolan GP, Perez OD. Analysis of protein phosphorylation and cellular signaling events by flow cytometry: techniques and clinical applications. Clin Immunol 2004; 110:206-21. [PMID: 15047199 DOI: 10.1016/j.clim.2003.11.009] [Citation(s) in RCA: 264] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Accepted: 11/10/2003] [Indexed: 01/16/2023]
Abstract
Analysis of protein phosphorylation with flow cytometric techniques has emerged as a powerful tool in the field of immunological signaling, allowing cellular subsets in complex populations to be analyzed accurately and rapidly. In this review, we examine the development of phospho-epitope, or phospho-specific, flow cytometry and the premises upon which the technique is based. Phospho-specific flow cytometry is compared to traditional biochemical methods, and its advantages, such as single cell analysis, multiparameter data acquisition, rapid protocols, and the ability to analyze rare cell subsets, are detailed. We also discuss the many technical considerations that must be addressed when developing new antibodies or analyzing new epitopes including antigen accessibility, stability of the phospho-epitope, fluorophore selection, surface phenotype integrity, and antibody suitability for staining epitopes inside fixed and permeabilized cells. The methods that have been used to date are described in light of these technical considerations. The importance of developing bioinformatic platforms in parallel with these techniques is emphasized due to the large, multiparameter datasets that are rapidly accumulated and which require more efficient data viewing and complex clustering methods than currently available for flow cytometric data. Finally, we discuss the potential clinical applications of phospho-specific flow cytometry in analyzing immune cell development and antigen-specific immune responses, as well as pharmacodynamic profiling of disease states or drug efficacy and specificity against particular signaling proteins.
Collapse
Affiliation(s)
- Peter O Krutzik
- Department of Molecular Pharmacology, School of Medicine, Stanford University, CA 94305, USA
| | | | | | | |
Collapse
|