1
|
Wessel RE, Dolatshahi S. Regulators of placental antibody transfer through a modeling lens. Nat Immunol 2024; 25:2024-2036. [PMID: 39379658 DOI: 10.1038/s41590-024-01971-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024]
Abstract
Infants are vulnerable to infections owing to a limited ability to mount a humoral immune response and their tolerogenic immune phenotype, which has impeded the success of newborn vaccination. Transplacental transfer of IgG from mother to fetus provides crucial protection in the first weeks of life, and maternal immunization has recently been implemented as a public health strategy to protect newborns against serious infections. Despite their early success, current maternal vaccines do not provide comparable protection across pregnancies with varying gestational lengths and placental and maternal immune features, and they do not account for the dynamic interplay between the maternal immune response and placental transfer. Moreover, progress toward the rational design of maternal vaccines has been hindered by inadequacies of existing experimental models and safety challenges of investigating longitudinal dynamics of IgG transfer in pregnant humans. Alternatively, in silico mechanistic models are a logical framework to disentangle the processes regulating placental antibody transfer. This Review synthesizes current literature through a mechanistic modeling lens to identify placental and maternal regulators of antibody transfer, their clinical covariates, and knowledge gaps to guide future research. We also describe opportunities to use integrated modeling and experimental approaches toward the rational design of vaccines against existing and emerging neonatal pathogen threats.
Collapse
Affiliation(s)
- Remziye E Wessel
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Virginia, Charlottesville, VA, USA
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Virginia, Charlottesville, VA, USA.
- Carter Immunology Center, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Chiang CJ, Hsu WL, Su MT, Ko WC, Hsu KF, Tsai PY. Impact of Antenatal SARS-CoV-2 Exposure on SARS-CoV-2 Neutralization Potency. Vaccines (Basel) 2024; 12:164. [PMID: 38400147 PMCID: PMC10892697 DOI: 10.3390/vaccines12020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
A pregnancy booster dose significantly reduces the risk and severity of COVID-19, and it is widely recommended. A prospective cohort study was conducted to compare the transplacental passage of maternal antibodies from vaccination or infection during three trimesters against both the vaccine-targeted Wuhan strain and the Omicron strain of SARS-CoV-2. Maternal-infant dyads from vaccinated mothers were collected between 6 June 2022 and 20 September 2022. We analyzed 38 maternal-infant dyads from mothers who had been infected with COVID-19 and 37 from mothers without any previous infection. Pregnant women who received their last COVID-19 vaccine dose in the third trimester exhibited the highest anti-spike protein antibody levels and neutralizing potency against both the Wuhan strain and Omicron BA.2 variant in their maternal and cord plasma. Both second- and third-trimester vaccination could lead to a higher level of neutralization against the Wuhan and Omicron strains. COVID-19 infection had a negative effect on the transplacental transfer ratio of SARS-CoV-2 antibodies. A booster dose during the second or third trimester is encouraged for the maximum transplacental transfer of humoral protection against COVID-19 for infants.
Collapse
Affiliation(s)
- Chia-Jung Chiang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Wei-Lun Hsu
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
- Center for Infection Control, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Keng-Fu Hsu
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704302, Taiwan
| |
Collapse
|
3
|
Gao C, Chen Q, Hao X, Wang Q. Immunomodulation of Antibody Glycosylation through the Placental Transfer. Int J Mol Sci 2023; 24:16772. [PMID: 38069094 PMCID: PMC10705935 DOI: 10.3390/ijms242316772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Establishing an immune balance between the mother and fetus during gestation is crucial, with the placenta acting as the epicenter of immune tolerance. The placental transfer of antibodies, mainly immunoglobulin G (IgG), is critical in protecting the developing fetus from infections. This review looks at how immunomodulation of antibody glycosylation occurs during placental transfer and how it affects fetal health. The passage of maternal IgG antibodies through the placental layers, including the syncytiotrophoblast, stroma, and fetal endothelium, is discussed. The effect of IgG subclass, glycosylation, concentration, maternal infections, and antigen specificity on antibody transfer efficiency is investigated. FcRn-mediated IgG transport, influenced by pH-dependent binding, is essential for placental transfer. Additionally, this review delves into the impact of glycosylation patterns on antibody functionality, considering both protective and pathological effects. Factors affecting the transfer of protective antibodies, such as maternal vaccination, are discussed along with reducing harmful antibodies. This in-depth examination of placental antibody transfer and glycosylation provides insights into improving neonatal immunity and mitigating the effects of maternal autoimmune and alloimmune conditions.
Collapse
Affiliation(s)
| | | | | | - Qiushi Wang
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
4
|
Wessel RE, Dolatshahi S. Quantitative mechanistic model reveals key determinants of placental IgG transfer and informs prenatal immunization strategies. PLoS Comput Biol 2023; 19:e1011109. [PMID: 37934786 PMCID: PMC10656024 DOI: 10.1371/journal.pcbi.1011109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 11/17/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023] Open
Abstract
Transplacental antibody transfer is crucially important in shaping neonatal immunity. Recently, prenatal maternal immunization has been employed to boost pathogen-specific immunoglobulin G (IgG) transfer to the fetus. Multiple factors have been implicated in antibody transfer, but how these key regulators work together to elicit selective transfer is pertinent to engineering vaccines for mothers to optimally immunize their newborns. Here, we present the first quantitative mechanistic model to uncover the determinants of placental antibody transfer and inform personalized immunization approaches. We identified placental FcγRIIb expressed by endothelial cells as a limiting factor in receptor-mediated transfer, which plays a key role in promoting preferential transport of subclasses IgG1, IgG3, and IgG4, but not IgG2. Integrated computational modeling and in vitro experiments reveal that IgG subclass abundance, Fc receptor (FcR) binding affinity, and FcR abundance in syncytiotrophoblasts and endothelial cells contribute to inter-subclass competition and potentially inter- and intra-patient antibody transfer heterogeneity. We developed an in silico prenatal vaccine testbed by combining a computational model of maternal vaccination with this placental transfer model using the tetanus, diphtheria, and acellular pertussis (Tdap) vaccine as a case study. Model simulations unveiled precision prenatal immunization opportunities that account for a patient's anticipated gestational length, placental size, and FcR expression by modulating vaccine timing, dosage, and adjuvant. This computational approach provides new perspectives on the dynamics of maternal-fetal antibody transfer in humans and potential avenues to optimize prenatal vaccinations that promote neonatal immunity.
Collapse
Affiliation(s)
- Remziye E. Wessel
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
5
|
Erdogan RR, Dolatshahi S. Quantitative mechanistic model reveals key determinants of placental IgG transfer and informs prenatal immunization strategies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537285. [PMID: 37131700 PMCID: PMC10153162 DOI: 10.1101/2023.04.18.537285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Transplacental antibody transfer is crucially important in shaping neonatal immunity. Recently, prenatal maternal immunization has been employed to boost pathogen-specific immunoglobulin G (IgG) transfer to the fetus. Multiple factors have been implicated in antibody transfer, but how these key dynamic regulators work together to elicit the observed selectivity is pertinent to engineering vaccines for mothers to optimally immunize their newborns. Here, we present the first quantitative mechanistic model to uncover the determinants of placental antibody transfer and inform personalized immunization approaches. We identified placental FcγRIIb expressed by endothelial cells as a limiting factor in receptor-mediated transfer, which plays a key role in promoting preferential transport of subclasses IgG1, IgG3, and IgG4, but not IgG2. Integrated computational modeling and in vitro experiments reveal that IgG subclass abundance, Fc receptor (FcR) binding affinity, and FcR abundance in syncytiotrophoblasts and endothelial cells contribute to inter-subclass competition and potentially inter- and intra-patient antibody transfer heterogeneity. We developed an in silico prenatal vaccine testbed by combining a computational model of maternal vaccination with this placental transfer model using the tetanus, diphtheria, and acellular pertussis (Tdap) vaccine as a case study. Model simulations unveiled precision prenatal immunization opportunities that account for a patient's anticipated gestational length, placental size, and FcR expression by modulating vaccine timing, dosage, and adjuvant. This computational approach provides new perspectives on the dynamics of maternal-fetal antibody transfer in humans and potential avenues to optimize prenatal vaccinations that promote neonatal immunity.
Collapse
Affiliation(s)
- Remziye R Erdogan
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, 22908
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, 22908
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, 22908
| |
Collapse
|
6
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
7
|
Dolatshahi S, Butler AL, Pou C, Henckel E, Bernhardsson AK, Gustafsson A, Bohlin K, Shin SA, Lauffenburger DA, Brodin P, Alter G. Selective transfer of maternal antibodies in preterm and fullterm children. Sci Rep 2022; 12:14937. [PMID: 36056073 PMCID: PMC9440225 DOI: 10.1038/s41598-022-18973-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 08/23/2022] [Indexed: 01/11/2023] Open
Abstract
Preterm newborns are more likely to suffer from infectious diseases at birth compared to children delivered at term. Whether this is due to compromised cellular, humoral, or organ-specific development remains unclear. To begin to define whether maternal-fetal antibody transfer profiles differ across preterm (PT) and fullterm (FT) infants, the overall quantity and functional quality of an array of 24 vaccine-, endemic pathogen-, and common antigen-specific antibodies were assessed across a cohort of 11 PT and 12 term-delivered maternal:infant pairs from birth through week 12. While total IgG levels to influenza, pneumo, measles, rubella, EBV, and RSV were higher in FT newborns, selective Fc-receptor binding antibodies was noted in PT newborns. In fact, near equivalent antibody-effector functions were observed across PT and FT infants, despite significant quantitative differences in transferred antibody levels. Moreover, temporal transfer analysis revealed the selective early transfer of FcRn, FcγR2, and FcγR3 binding antibodies, pointing to differential placental sieving mechanisms across gestation. These data point to selectivity in placental transfer at distinct gestational ages, to ensure that children are endowed with the most robust humoral immunity even if born preterm.
Collapse
Affiliation(s)
- Sepideh Dolatshahi
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, USA
| | | | - Christian Pou
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Henckel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anna Karin Bernhardsson
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anna Gustafsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Kajsa Bohlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Sally A Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering and Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
- Karolinska University Hospital, Stockholm, Sweden.
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Estrogen-Driven Changes in Immunoglobulin G Fc Glycosylation. EXPERIENTIA. SUPPLEMENTUM 2021. [PMID: 34687016 DOI: 10.1007/978-3-030-76912-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Glycosylation within the immunoglobulin G (IgG) Fc region modulates its ability to engage complement and Fc receptors, affording the opportunity to fine-tune effector functions. Mechanisms regulating IgG Fc glycans remain poorly understood. Changes accompanying menarche, menopause, and pregnancy have long implicated hormonal factors. Intervention studies now confirm that estrogens enhance IgG Fc galactosylation, in females and also in males, defining the first pathway modulating Fc glycans and thereby a new link between sex and immunity. This mechanism may participate in fetal-maternal immunity, antibody-mediated inflammation, and other aspects of age- and sex-specific immune function. Here we review the changes affecting the IgG Fc glycome from childhood through old age, the evidence establishing a role for estrogens, and research directions to uncover associated mechanisms that may inform therapeutic intervention.
Collapse
|
9
|
Yockey LJ, Lucas C, Iwasaki A. Contributions of maternal and fetal antiviral immunity in congenital disease. Science 2020; 368:608-612. [PMID: 32381717 DOI: 10.1126/science.aaz1960] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
Abstract
Viral infections during pregnancy can have devastating consequences on pregnancy outcomes, fetal development, and maternal health. In this review, we examine fetal and maternal immune defense mechanisms that mediate resistance against viral infections and discuss the range of syndromes that ensue when such mechanisms fail, from fetal developmental defects to establishment of chronic infection. Further, we highlight the role of maternal immune activation, or uncontrolled inflammation triggered by viral infections during pregnancy, and its potential downstream pathological effects, including tissue damage and fetal demise. Insights into the respective contributions of direct viral toxicity versus fetal and maternal immune responses that underlie the pathogenesis of congenital disease will guide future treatment strategies.
Collapse
Affiliation(s)
- Laura J Yockey
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.,Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carolina Lucas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06512, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
10
|
Abl family tyrosine kinases govern IgG extravasation in the skin in a murine pemphigus model. Nat Commun 2019; 10:4432. [PMID: 31570755 PMCID: PMC6769004 DOI: 10.1038/s41467-019-12232-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/28/2019] [Indexed: 01/02/2023] Open
Abstract
The pathway of homeostatic IgG extravasation is not fully understood, in spite of its importance for the maintenance of host immunity, the management of autoantibody-mediated disorders, and the use of antibody-based biologics. Here we show in a murine model of pemphigus, a prototypic cutaneous autoantibody-mediated disorder, that blood-circulating IgG extravasates into the skin in a time- and dose-dependent manner under homeostatic conditions. This IgG extravasation is unaffected by depletion of Fcγ receptors, but is largely attenuated by specific ablation of dynamin-dependent endocytic vesicle formation in blood endothelial cells (BECs). Among dynamin-dependent endocytic vesicles, IgG co-localizes well with caveolae in cultured BECs. An Abl family tyrosine kinase inhibitor imatinib, which reduces caveolae-mediated endocytosis, impairs IgG extravasation in the skin and attenuates the murine pemphigus manifestations. Our study highlights the kinetics of IgG extravasation in vivo, which might be a clue to understand the pathological mechanism of autoantibody-mediated autoimmune disorders. How antibody reaches tissues from circulation is critical for understanding antibody-mediated immunity. Here the authors show that IgG extravasation in the skin is mediated by endothelial caveolin transport independently of FcR, and is targetable by imatinib, which reduces IgG-dependent pathology in a mouse model of pemphigus.
Collapse
|
11
|
Surface-Functionalized Nanoparticles as Efficient Tools in Targeted Therapy of Pregnancy Complications. Int J Mol Sci 2019; 20:ijms20153642. [PMID: 31349643 PMCID: PMC6695948 DOI: 10.3390/ijms20153642] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/02/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022] Open
Abstract
Minimizing exposure of the fetus to medication and reducing adverse off-target effects in the mother are the primary challenges in developing novel drugs to treat pregnancy complications. Nanomedicine has introduced opportunities for the development of novel platforms enabling targeted delivery of drugs in pregnancy. This review sets out to discuss the advances and potential of surface-functionalized nanoparticles in the targeted therapy of pregnancy complications. We first describe the human placental anatomy, which is fundamental for developing placenta-targeted therapy, and then we review current knowledge of nanoparticle transplacental transport mechanisms. Meanwhile, recent surface-functionalized nanoparticles for targeting the uterus and placenta are examined. Indeed, surface-functionalized nanoparticles could help prevent transplacental passage and promote placental-specific drug delivery, thereby enhancing efficacy and improving safety. We have achieved promising results in targeting the placenta via placental chondroitin sulfate A (plCSA), which is exclusively expressed in the placenta, using plCSA binding peptide (plCSA-BP)-decorated nanoparticles. Others have also focused on using placenta- and uterus-enriched molecules as targets to deliver therapeutics via surface-functionalized nanoparticles. Additionally, we propose that placenta-specific exosomes and surface-modified exosomes might be potential tools in the targeted therapy of pregnancy complications. Altogether, surface-functionalized nanoparticles have great potential value as clinical tools in the targeted therapy of pregnancy complications.
Collapse
|
12
|
Pyzik M, Sand KMK, Hubbard JJ, Andersen JT, Sandlie I, Blumberg RS. The Neonatal Fc Receptor (FcRn): A Misnomer? Front Immunol 2019; 10:1540. [PMID: 31354709 PMCID: PMC6636548 DOI: 10.3389/fimmu.2019.01540] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies are essential components of an adaptive immune response. Immunoglobulin G (IgG) is the most common type of antibody found in circulation and extracellular fluids. Although IgG alone can directly protect the body from infection through the activities of its antigen binding region, the majority of IgG immune functions are mediated via proteins and receptors expressed by specialized cell subsets that bind to the fragment crystallizable (Fc) region of IgG. Fc gamma (γ) receptors (FcγR) belong to a broad family of proteins that presently include classical membrane-bound surface receptors as well as atypical intracellular receptors and cytoplasmic glycoproteins. Among the atypical FcγRs, the neonatal Fc receptor (FcRn) has increasingly gained notoriety given its intimate influence on IgG biology and its ability to also bind to albumin. FcRn functions as a recycling or transcytosis receptor that is responsible for maintaining IgG and albumin in the circulation, and bidirectionally transporting these two ligands across polarized cellular barriers. More recently, it has been appreciated that FcRn acts as an immune receptor by interacting with and facilitating antigen presentation of peptides derived from IgG immune complexes (IC). Here we review FcRn biology and focus on newer advances including how emerging FcRn-targeted therapies may affect the immune responses to IgG and IgG IC.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Kine M K Sand
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jonathan J Hubbard
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Digestive Diseases Center, Boston, MA, United States
| |
Collapse
|
13
|
Jennewein MF, Goldfarb I, Dolatshahi S, Cosgrove C, Noelette FJ, Krykbaeva M, Das J, Sarkar A, Gorman MJ, Fischinger S, Boudreau CM, Brown J, Cooperrider JH, Aneja J, Suscovich TJ, Graham BS, Lauer GM, Goetghebuer T, Marchant A, Lauffenburger D, Kim AY, Riley LE, Alter G. Fc Glycan-Mediated Regulation of Placental Antibody Transfer. Cell 2019; 178:202-215.e14. [PMID: 31204102 PMCID: PMC6741440 DOI: 10.1016/j.cell.2019.05.044] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/07/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023]
Abstract
Despite the worldwide success of vaccination, newborns remain vulnerable to infections. While neonatal vaccination has been hampered by maternal antibody-mediated dampening of immune responses, enhanced regulatory and tolerogenic mechanisms, and immune system immaturity, maternal pre-natal immunization aims to boost neonatal immunity via antibody transfer to the fetus. However, emerging data suggest that antibodies are not transferred equally across the placenta. To understand this, we used systems serology to define Fc features associated with antibody transfer. The Fc-profile of neonatal and maternal antibodies differed, skewed toward natural killer (NK) cell-activating antibodies. This selective transfer was linked to digalactosylated Fc-glycans that selectively bind FcRn and FCGR3A, resulting in transfer of antibodies able to efficiently leverage innate immune cells present at birth. Given emerging data that vaccination may direct antibody glycosylation, our study provides insights for the development of next-generation maternal vaccines designed to elicit antibodies that will most effectively aid neonates.
Collapse
Affiliation(s)
| | - Ilona Goldfarb
- Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sepideh Dolatshahi
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cormac Cosgrove
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Marina Krykbaeva
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jishnu Das
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aniruddh Sarkar
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthew J Gorman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | | | - Joelle Brown
- Gastroenterology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Jasneet Aneja
- Gastroenterology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Todd J Suscovich
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, Bethesda, MD 20892, USA
| | - Georg M Lauer
- Gastroenterology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tessa Goetghebuer
- Department of Pediatrics, Hôpital Saint-Pierre, Brussels 1000, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi 6041, Belgium
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; MIT Center for Gynepathology Research, Cambridge, MA 02139, USA
| | - Arthur Y Kim
- Division of Infectious Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Laura E Riley
- Department of Obstetrics and Gynecology, New York Presbyterian/Weill Cornell Medical Center, New York, NY 10065, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
14
|
Goller SS, Markert UR, Fröhlich K. Trastuzumab in the Treatment of Pregnant Breast Cancer Patients - an Overview of the Literature. Geburtshilfe Frauenheilkd 2019; 79:618-625. [PMID: 31217630 PMCID: PMC6570610 DOI: 10.1055/a-0880-9295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 02/08/2019] [Accepted: 03/20/2019] [Indexed: 01/11/2023] Open
Abstract
Breast cancer is one of the most common malignancies which appear during pregnancy. Since women are increasingly not giving birth until they are at a more advanced age, it can be assumed that the incidence of pregnancy-related breast cancers will continue to increase in the future. Because of pregnancy-induced changes and conservative diagnosis, these carcinomas are frequently not detected until they are at an advanced stage and thus generally require systemic adjuvant therapy. The available data on optimal chemotherapeutic management are limited. Particularly for the use of the target agent trastuzumab which could crucially contribute to improving the prognosis in the therapy of HER2-overexpressing breast cancer in non-pregnant women, there is a lack of definitive information regarding the profile of action and safety in pregnancy as well as with regard to any long-term effects on the child. Thirty-eight pregnancies on trastuzumab for the treatment of breast cancer were able to be analysed in the literature currently available. Information can be gained from this and conclusions can be drawn which can individualise and decisively improve therapeutic options in the future for the pregnant breast cancer patient.
Collapse
Affiliation(s)
- Sophia S Goller
- Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
| | - Udo R Markert
- Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
| | - Karolin Fröhlich
- Universitätsklinikum Jena, Klinik für Geburtsmedizin, Placenta-Labor, Jena, Germany
| |
Collapse
|
15
|
Kiskova T, Mytsko Y, Schepelmann M, Helmer H, Fuchs R, Miedl H, Wadsack C, Ellinger I. Expression of the neonatal Fc-receptor in placental-fetal endothelium and in cells of the placental immune system. Placenta 2019; 78:36-43. [PMID: 30955709 DOI: 10.1016/j.placenta.2019.02.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/21/2019] [Accepted: 02/22/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Starting from the second trimester of pregnancy, passive immunity is provided to the human fetus by transplacental transfer of maternal IgG. IgG transfer depends on the neonatal Fc receptor, FcRn. While FcRn localization in the placental syncytiotrophoblast (STB) has been demonstrated unequivocally, FcRn expression in placental-fetal endothelial cells (pFECs), which are part of the materno-fetal barrier, is still unclear. Therefore, this study aimed to elucidate the spatio-specific expression pattern of FcRn in placental tissue. METHODS FcRn expression was investigated by western blotting in term placentas and in isolated human placental arterial and venous endothelial cells (HPAEC, HPVEC) using a validated affinity-purified polyclonal anti-peptide antibody against the cytoplasmic tail of FcRn α-chain. In situ localization of FcRn and IgG was studied by immunofluorescence microscopy on tissue sections of healthy term placentas. RESULTS FcRn expression was demonstrated in placental vasculature particularly, in HPAEC, and HPVEC. FcRn was localized in cytokeratin 7+ STB and in CD31+ pFECs in terminal as well as stem villi in situ. Additionally, CD68+ placental macrophages exhibited FcRn expression in situ. Endogenous IgG partially co-localized with FcRn in STB, pFECs, and in placental macrophages. DISCUSSION Placental FcRn expression in endothelial cells and macrophages is analogous to the expression pattern in other organs. FcRn expression in pFECs suggests an involvement of FcRn in IgG transcytosis and/or participation in recycling/salvaging of maternal IgG present in the fetal circulation. FcRn expression in placental macrophages may account for recycling of monomeric IgG and/or processing and presentation of immune complexes.
Collapse
Affiliation(s)
- Terezia Kiskova
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Yuliya Mytsko
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Martin Schepelmann
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Hanns Helmer
- Division of Obstetrics and Feto-Maternal Medicine, University Department of Obstetrics and Gynaecology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Renate Fuchs
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Heidi Miedl
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Isabella Ellinger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
16
|
Martinez DR, Fouda GG, Peng X, Ackerman ME, Permar SR. Noncanonical placental Fc receptors: What is their role in modulating transplacental transfer of maternal IgG? PLoS Pathog 2018; 14:e1007161. [PMID: 30161231 PMCID: PMC6117057 DOI: 10.1371/journal.ppat.1007161] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- David R. Martinez
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Xinxia Peng
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Sallie R. Permar
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
17
|
Stapleton NM, Armstrong-Fisher SS, Andersen JT, van der Schoot CE, Porter C, Page KR, Falconer D, de Haas M, Williamson LM, Clark MR, Vidarsson G, Armour KL. Human IgG lacking effector functions demonstrate lower FcRn-binding and reduced transplacental transport. Mol Immunol 2018; 95:1-9. [PMID: 29367080 DOI: 10.1016/j.molimm.2018.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/07/2018] [Accepted: 01/10/2018] [Indexed: 11/24/2022]
Abstract
We have previously generated human IgG1 antibodies that were engineered for reduced binding to the classical Fcγ receptors (FcγRI-III) and C1q, thereby eliminating their destructive effector functions (constant region G1Δnab). In their potential use as blocking agents, favorable binding to the neonatal Fc receptor (FcRn) is important to preserve the long half-life typical of IgG. An ability to cross the placenta, which is also mediated, at least in part, by FcRn is desirable in some indications, such as feto-maternal alloimmune disorders. Here, we show that G1Δnab mutants retain pH-dependent binding to human FcRn but that the amino acid alterations reduce the affinity of the IgG1:FcRn interaction by 2.0-fold and 1.6-fold for the two antibodies investigated. The transport of the modified G1Δnab mutants across monolayers of human cell lines expressing FcRn was approximately 75% of the wild-type, except that no difference was observed with human umbilical vein endothelial cells. G1Δnab mutation also reduced transport in an ex vivo placenta model. In conclusion, we demonstrate that, although the G1Δnab mutations are away from the FcRn-binding site, they have long-distance effects, modulating FcRn binding and transcellular transport. Our findings have implications for the design of therapeutic human IgG with tailored effector functions.
Collapse
Affiliation(s)
- Nigel M Stapleton
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, Amsterdam, 1066 CX, The Netherlands
| | - Sylvia S Armstrong-Fisher
- RDI Clinical Transfusion Group, Scottish National Blood Transfusion Service, Foresterhill, Aberdeen, AB25 2ZW, UK; Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4950, Nydalen, Oslo, 0424, Norway; Centre for Immune Regulation and Department of Biosciences, University of Oslo, PO box 1041, Blindern, Oslo, 0316, Norway; Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Problemveien 7, 0315, Oslo, Norway
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, Amsterdam, 1066 CX, The Netherlands
| | - Charlene Porter
- Immunology Laboratory, Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, AB25 2ZB, UK
| | - Kenneth R Page
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Donald Falconer
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Masja de Haas
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, Amsterdam, 1066 CX, The Netherlands
| | - Lorna M Williamson
- Department of Haematology, University of Cambridge, UK; NHS Blood and Transplant, Long Road, Cambridge, CB2 2PT, UK
| | - Michael R Clark
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Plesmanlaan 125, Amsterdam, 1066 CX, The Netherlands.
| | - Kathryn L Armour
- Department of Haematology, University of Cambridge, UK; Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
18
|
Roghanian A, Stopforth RJ, Dahal LN, Cragg MS. New revelations from an old receptor: Immunoregulatory functions of the inhibitory Fc gamma receptor, FcγRIIB (CD32B). J Leukoc Biol 2018; 103:1077-1088. [PMID: 29406570 DOI: 10.1002/jlb.2mir0917-354r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The Fc gamma receptor IIB (FcγRIIB/CD32B) was generated million years ago during evolution. It is the sole inhibitory receptor for IgG, and has long been associated with the regulation of humoral immunity and innate immune homeostasis. However, new and surprising functions of FcγRIIB are emerging. In particular, FcγRIIB has been shown to perform unexpected activatory roles in both immune-signaling and monoclonal antibody (mAb) immunotherapy. Furthermore, although ITIM signaling is an integral part of FcγRIIB regulatory activity, it is now clear that inhibition/activation of immune responses can occur independently of the ITIM. In light of these new findings, we present an overview of the established and noncanonical functions of FcγRIIB and discuss how this knowledge might be exploited therapeutically.
Collapse
Affiliation(s)
- Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard J Stopforth
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
19
|
Fouda GG, Martinez DR, Swamy GK, Permar SR. The Impact of IgG transplacental transfer on early life immunity. Immunohorizons 2018; 2:14-25. [PMID: 29457151 PMCID: PMC5812294 DOI: 10.4049/immunohorizons.1700057] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Pediatric vaccines have significantly reduced infectious disease-related infant mortality, but as protective immunity often require several infant vaccine doses; maternally-acquired antibodies are critical to protect infants during the first months of life. Consequently, immunization of pregnant women is an important strategy not only to protect mothers from infection, but also to provide immunity to young infants. Nevertheless, maternal immunization can also negatively impact early life immunity. In fact, maternal antibodies can interfere with the development of infant immune responses, though it is unclear if such interference is clinically significant. Moreover, the transplacental transfer of maternal immunoglobulin therapeutics can be harmful to the fetus. Thus, the risk/benefit of maternal immunization for both the mother and the fetus should be carefully weighed. In addition, it is critical to fully understand the mechanisms by which IgG is transferred across the placenta in order to develop optimal maternal and infant immunization strategies.
Collapse
Affiliation(s)
- Genevieve G. Fouda
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710
| | - David R. Martinez
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 27710
| | - Geeta K. Swamy
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, 27710
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710
| |
Collapse
|
20
|
Wilcox CR, Holder B, Jones CE. Factors Affecting the FcRn-Mediated Transplacental Transfer of Antibodies and Implications for Vaccination in Pregnancy. Front Immunol 2017; 8:1294. [PMID: 29163461 PMCID: PMC5671757 DOI: 10.3389/fimmu.2017.01294] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/26/2017] [Indexed: 11/30/2022] Open
Abstract
At birth, neonates are particularly vulnerable to infection and transplacental transfer of immunoglobulin G (IgG) from mother to fetus provides crucial protection in the first weeks of life. Transcytosis of IgG occurs via binding with the neonatal Fc receptor (FcRn) in the placental synctiotrophoblast. As maternal vaccination becomes an increasingly important strategy for the protection of young infants, improving our understanding of transplacental transfer and the factors that may affect this will become increasingly important, especially in low-income countries where the burden of morbidity and mortality is highest. This review highlights factors of relevance to maternal vaccination that may modulate placental transfer—IgG subclass, glycosylation of antibody, total maternal IgG concentration, maternal disease, infant gestational age, and birthweight—and outlines the conflicting evidence and questions that remain regarding the complexities of these relationships. Furthermore, the intricacies of the Ab–FcRn interaction remain poorly understood and models that may help address future research questions are described.
Collapse
Affiliation(s)
- Christopher R Wilcox
- National Institute of Health Research Wellcome Trust Clinical Research Facility, Southampton, United Kingdom
| | - Beth Holder
- Paediatrics Section, Division of Infectious Diseases, Centre for International Child Health, Imperial College London, London, United Kingdom
| | - Christine E Jones
- Faculty of Medicine, Institute for Life Sciences, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
21
|
Jennewein MF, Abu-Raya B, Jiang Y, Alter G, Marchant A. Transfer of maternal immunity and programming of the newborn immune system. Semin Immunopathol 2017; 39:605-613. [PMID: 28971246 DOI: 10.1007/s00281-017-0653-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
Abstract
As placental mammals, the pregnant women and the fetus have intense and prolonged interactions during gestation. There is increasing evidence that multiple molecular as well as cellular components originating in pregnant women are transferred to the fetus. The transfer of maternal antibodies has long been recognized as a central component of newborn immunity against pathogens. More recent studies indicate that inflammatory mediators, micronutrients, microbial products and maternal cells are transferred in utero and influence the fetal immune system. Together, these multiple signals are likely to form a complex network of interactions that program the neonatal immune system and tune its homeostatic regulation. Maternal disorders, in particular infectious diseases, modify these signals and may thereby alter immunity in early life. Understanding maternal programming of the newborn immune system could provide a basis for interventions promoting child health.
Collapse
Affiliation(s)
| | - Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital, Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Yiwei Jiang
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium.
| |
Collapse
|
22
|
Ono S, Egawa G, Kabashima K. Regulation of blood vascular permeability in the skin. Inflamm Regen 2017; 37:11. [PMID: 29259710 PMCID: PMC5725833 DOI: 10.1186/s41232-017-0042-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/25/2017] [Indexed: 12/31/2022] Open
Abstract
Regulation of blood vessel permeability is essential for the homeostasis of peripheral tissues. This regulation controls the trafficking of plasma contents, including water, vitamins, ions, hormones, cytokines, amyloids, lipoproteins, carrier proteins, and immunoglobulins. The properties of blood vessels vary among tissues based on their structural differences: continuous, fenestrated, or sinusoidal. These three types of blood vessels have different charge and size barrier properties. The anionic luminal glycocalyx layer on endothelial cells establishes the "charge barrier" that repels the attachment of negatively charged blood cells and plasma molecules. In contrast, the "size barrier" of blood vessels largely relies on the interendothelial junctions (IEJs) between endothelial cells, which define the paracellular permeability. As in most peripheral tissues, blood capillaries in the skin are composed of continuous and/or fenestrated blood vessels that have relatively tighter IEJs compared to those in the internal organs. Small vesicles in the capillary endothelium were discovered in the 1950s, and studies have since confirmed that blood endothelial cells transport the plasma contents by endocytosis and subsequent transcytosis and exocytosis-this process is called transcellular permeability. The permeability of blood vessels is highly variable as a result of intrinsic and extrinsic factors. It is significantly elevated upon tissue inflammations as a result of disabled IEJs and increased paracellular permeability due to inflammatory mediators. An increase in transcellular permeability during inflammation has also been postulated. Here, we provide an overview of the general properties of vascular permeability based on our recent observations of murine skin inflammation models, and we discuss its physiological significance in peripheral homeostasis.
Collapse
Affiliation(s)
- Sachiko Ono
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
| | - Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara, Sakyo, Kyoto, 606-8507 Japan
- Singapore Immunology Network (SIgN) and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
23
|
Heidl S, Ellinger I, Niederberger V, Waltl EE, Fuchs R. Localization of the human neonatal Fc receptor (FcRn) in human nasal epithelium. PROTOPLASMA 2016; 253:1557-1564. [PMID: 26634928 DOI: 10.1007/s00709-015-0918-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/25/2015] [Indexed: 06/05/2023]
Abstract
The airway epithelium is a central player in the defense against pathogens including efficient mucociliary clearance and secretion of immunoglobulins, mainly polymeric IgA, but also IgG. Pulmonary administration of therapeutic antibodies on one hand, and intranasal immunization on the other, are powerful tools to treat airway infections. In either case, the airway epithelium is the primary site of antibody transfer. In various epithelia, bi-polar transcytosis of IgG and IgG immune complexes is mediated by the human neonatal Fc receptor, FcRn, but FcRn expression in the nasal epithelium had not been demonstrated, so far. We prepared affinity-purified antibodies against FcRn α-chain and confirmed their specificity by Western blotting and immunofluorescence microscopy. These antibodies were used to study the localization of FcRn α-chain in fixed nasal tissue. We here demonstrate for the first time that ciliated epithelial cells, basal cells, gland cells, and endothelial cells in the underlying connective tissue express the receptor. A predominant basolateral steady state distribution of the receptor was observed in ciliated epithelial as well as in gland cells. Co-localization of FcRn α-chain with IgG or with early sorting endosomes (EEA1-positive) but not with late endosomes/lysosomes (LAMP-2-positive) in ciliated cells was observed. This is indicative for the presence of the receptor in the recycling/transcytotic pathway but not in compartments involved in lysosomal degradation supporting the role of FcRn in IgG transcytosis in the nasal epithelium.
Collapse
Affiliation(s)
- Sara Heidl
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Isabella Ellinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Verena Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Eva E Waltl
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Renate Fuchs
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
24
|
Scavenger receptor B1, the HDL receptor, is expressed abundantly in liver sinusoidal endothelial cells. Sci Rep 2016; 6:20646. [PMID: 26865459 PMCID: PMC4749959 DOI: 10.1038/srep20646] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/07/2016] [Indexed: 02/08/2023] Open
Abstract
Cholesterol from peripheral tissue, carried by HDL, is metabolized in the liver after uptake by the HDL receptor, SR-B1. Hepatocytes have long been considered the only liver cells expressing SR-B1; however, in this study we describe two disparate immunofluorescence (IF) experiments that suggest otherwise. Using high-resolution confocal microscopy employing ultrathin (120 nm) sections of mouse liver, improving z-axis resolution, we identified the liver sinusoidal endothelial cells (LSEC), marked by FcγRIIb, as the cell within the liver expressing abundant SR-B1. In contrast, the hepatocyte, identified with β-catenin, expressed considerably weaker levels, although optical resolution of SR-B1 was inadequate. Thus, we moved to a different IF strategy, first separating dissociated liver cells by gradient centrifugation into two portions, hepatocytes (parenchymal cells) and LSEC (non-parenchymal cells). Characterizing both portions for the cellular expression of SR-B1 by flow cytometry, we found that LSEC expressed considerable amounts of SR-B1 while in hepatocytes SR-B1 expression was barely perceptible. Assessing mRNA of SR-B1 by real time PCR we found messenger expression in LSEC to be about 5 times higher than in hepatocytes.
Collapse
|
25
|
Foss S, Grevys A, Sand KMK, Bern M, Blundell P, Michaelsen TE, Pleass RJ, Sandlie I, Andersen JT. Enhanced FcRn-dependent transepithelial delivery of IgG by Fc-engineering and polymerization. J Control Release 2015; 223:42-52. [PMID: 26718855 DOI: 10.1016/j.jconrel.2015.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/14/2015] [Accepted: 12/19/2015] [Indexed: 01/28/2023]
Abstract
Monoclonal IgG antibodies (Abs) are used extensively in the clinic to treat cancer and autoimmune diseases. In addition, therapeutic proteins are genetically fused to the constant Fc part of IgG. In both cases, the Fc secures a long serum half-life and favourable pharmacokinetics due to its pH-dependent interaction with the neonatal Fc receptor (FcRn). FcRn also mediates transport of intact IgG across polarized epithelial barriers, a pathway that is attractive for delivery of Fc-containing therapeutics. So far, no study has thoroughly compared side-by-side how IgG and different Fc-fusion formats are transported across human polarizing epithelial cells. Here, we used an in vitro cellular transport assay based on the human polarizing epithelial cell line (T84) in which both IgG1 and Fc-fusions were transported in an FcRn-dependent manner. Furthermore, we found that the efficacy of transport was dependent on the format. We demonstrate that transepithelial delivery could be enhanced by Fc-engineering for improved FcRn binding as well as by Fc-polymerization. In both cases, transport was driven by pH-dependent binding kinetics and the pH at the luminal side. Hence, efficient transcellular delivery of IgG-based drugs across human epithelial cells requires optimal pH-dependent FcRn binding that can be manipulated by avidity and Fc-engineering, factors that should inspire the design of future therapeutics targeted for transmucosal delivery.
Collapse
Affiliation(s)
- Stian Foss
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Algirdas Grevys
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Kine Marita Knudsen Sand
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Malin Bern
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Pat Blundell
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Terje E Michaelsen
- Department of Bacteriology and Immunology, Norwegian Institute of Public Health, Oslo, Norway; Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Richard J Pleass
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Inger Sandlie
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR), Department of Biosciences, University of Oslo, N-0316, Oslo, Norway; Department of Immunology and CIR, Oslo University Hospital, Rikshospitalet, University of Oslo, N-0372, Oslo, Norway.
| |
Collapse
|
26
|
Anderson CL, Ganesan LP, Robinson JM. The biology of the classical Fcγ receptors in non-hematopoietic cells. Immunol Rev 2015; 268:236-40. [DOI: 10.1111/imr.12335] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Takizawa T, Powell RD, Hainfeld JF, Robinson JM. FluoroNanogold: an important probe for correlative microscopy. J Chem Biol 2015; 8:129-42. [PMID: 26884817 PMCID: PMC4744603 DOI: 10.1007/s12154-015-0145-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 07/24/2015] [Indexed: 10/23/2022] Open
Abstract
Correlative microscopy is a powerful imaging approach that refers to observing the same exact structures within a specimen by two or more imaging modalities. In biological samples, this typically means examining the same sub-cellular feature with different imaging methods. Correlative microscopy is not restricted to the domains of fluorescence microscopy and electron microscopy; however, currently, most correlative microscopy studies combine these two methods, and in this review, we will focus on the use of fluorescence and electron microscopy. Successful correlative fluorescence and electron microscopy requires probes, or reporter systems, from which useful information can be obtained with each of the imaging modalities employed. The bi-functional immunolabeling reagent, FluoroNanogold, is one such probe that provides robust signals in both fluorescence and electron microscopy. It consists of a gold cluster compound that is visualized by electron microscopy and a covalently attached fluorophore that is visualized by fluorescence microscopy. FluoroNanogold has been an extremely useful labeling reagent in correlative microscopy studies. In this report, we present an overview of research using this unique probe.
Collapse
Affiliation(s)
| | - Richard D. Powell
- />Nanoprobes, Incorporated, 95 Horseblock Road, Unit 1, Yaphank, NY 11980-9710 USA
| | - James F. Hainfeld
- />Nanoprobes, Incorporated, 95 Horseblock Road, Unit 1, Yaphank, NY 11980-9710 USA
| | - John M. Robinson
- />Department of Physiology and Cell Biology, Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
28
|
Adar T, Grisaru-Granovsky S, Ben Ya'acov A, Goldin E, Bar-Gil Shitrit A. Pregnancy and the Immune System: General Overview and the Gastroenterological Perspective. Dig Dis Sci 2015; 60:2581-9. [PMID: 25947331 DOI: 10.1007/s10620-015-3683-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/22/2015] [Indexed: 12/11/2022]
Abstract
Pregnancy represents a unique immune tolerant condition that cannot be attributed merely to generalized immunosuppression. A variety of mechanisms have been described, ranging from the non-self recognition, immunomodulation of specific inflammatory cell populations and a Th2-directed shift of the immune response, which are mediated by both localized and systemic mediators. Furthermore, an inflammatory response directed toward the conceptus is no longer considered an obligatory deleterious response; instead, it is considered an important factor that is necessary for normal growth and development. These immunomodulatory changes during pregnancy may also affect concurrent conditions and alter the course of inflammatory diseases. Herein, we review the main immunomodulatory changes that occur during pregnancy and their effect on coexisting inflammatory conditions, with a specific focus on gastrointestinal disorders.
Collapse
Affiliation(s)
- Tomer Adar
- Digestive Disease Institute, Shaare Zedek Medical Center, Affiliated with the School of Medicine in the Hebrew University of Jerusalem, 12 Bayit St., 91031, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
29
|
Ishikawa T, Takizawa T, Iwaki J, Mishima T, Ui-Tei K, Takeshita T, Matsubara S, Takizawa T. Fc gamma receptor IIb participates in maternal IgG trafficking of human placental endothelial cells. Int J Mol Med 2015; 35:1273-89. [PMID: 25778799 PMCID: PMC4380207 DOI: 10.3892/ijmm.2015.2141] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/17/2015] [Indexed: 12/21/2022] Open
Abstract
The human placental transfer of maternal IgG is crucial for fetal and newborn immunity. Low-affinity immunoglobulin gamma Fc region receptor IIb2 (FCGR2B2 or FcγRIIb2) is exclusively expressed in an IgG-containing, vesicle-like organelle (the FCGR2B2 compartment) in human placental endothelial cells; thus, we hypothesized that the FCGR2B2 compartment functions as an IgG transporter. In this study, to examine this hypothesis, we performed in vitro bio-imaging analysis of IgG trafficking by FCGR2B2 compartments using human umbilical vein endothelial cells transfected with a plasmid vector containing enhanced GFP-tagged FCGR2B2 (pFCGR2B2-EGFP). FCGR2B2-EGFP signals were detected as intracellular vesicular structures similar to FCGR2B2 compartments in vivo. The internalization and transcytosis of IgG was significantly higher in the pFCGR2B2-EGFP-transfected cells than in the mock-transfected cells, and the majority of the internalized IgG was co-localized with the FCGR2B2-EGFP signals. Furthermore, we isolated FCGR2B2 compartments from the human placenta and found that the Rab family of proteins [RAS-related protein Rab family (RABs)] were associated with FCGR2B2 compartments. Among the RABs, RAB3D was expressed predominantly in placental endothelial cells. The downregulation of RAB3D by small interfering RNA (siRNA) resulted in a marked reduction in the FCGR2B2-EGFP signals at the cell periphery. Taken together, these findings suggest that FCGR2B2 compartments participate in the transcytosis of maternal IgG across the human placental endothelium and that RAB3D plays a role in regulating the intracellular dynamics of FCGR2B2 compartments.
Collapse
Affiliation(s)
- Tomoko Ishikawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Takami Takizawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Jun Iwaki
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Takuya Mishima
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | - Kumiko Ui-Tei
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Toshiyuki Takeshita
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Shigeki Matsubara
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Toshihiro Takizawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
30
|
Sand KMK, Bern M, Nilsen J, Noordzij HT, Sandlie I, Andersen JT. Unraveling the Interaction between FcRn and Albumin: Opportunities for Design of Albumin-Based Therapeutics. Front Immunol 2015; 5:682. [PMID: 25674083 PMCID: PMC4306297 DOI: 10.3389/fimmu.2014.00682] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/17/2014] [Indexed: 01/08/2023] Open
Abstract
The neonatal Fc receptor (FcRn) was first found to be responsible for transporting antibodies of the immunoglobulin G (IgG) class from the mother to the fetus or neonate as well as for protecting IgG from intracellular catabolism. However, it has now become apparent that the same receptor also binds albumin and plays a fundamental role in homeostatic regulation of both IgG and albumin, as FcRn is expressed in many different cell types and organs at diverse body sites. Thus, to gain a complete understanding of the biological function of each ligand, and also their distribution in the body, an in-depth characterization of how FcRn binds and regulates the transport of both ligands is necessary. Importantly, such knowledge is also relevant when developing new drugs, as IgG and albumin are increasingly utilized in therapy. This review discusses our current structural and biological understanding of the relationship between FcRn and its ligands, with a particular focus on albumin and design of albumin-based therapeutics.
Collapse
Affiliation(s)
- Kine Marita Knudsen Sand
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Malin Bern
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jeannette Nilsen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway ; Institute of Clinical Medicine, University of Oslo , Oslo , Norway
| | - Hanna Theodora Noordzij
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation (CIR), University of Oslo , Oslo , Norway ; Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation (CIR), Oslo University Hospital Rikshospitalet , Oslo , Norway
| |
Collapse
|
31
|
Einarsdottir HK, Stapleton NM, Scherjon S, Andersen JT, Rispens T, van der Schoot CE, Vidarsson G. On the perplexingly low rate of transport of IgG2 across the human placenta. PLoS One 2014; 9:e108319. [PMID: 25251461 PMCID: PMC4177109 DOI: 10.1371/journal.pone.0108319] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/05/2014] [Indexed: 02/04/2023] Open
Abstract
The neonatal receptor, FcRn, mediates both serum half–life extension as well as active transport of maternal IgG to the fetus during pregnancy. Therefore, transport efficiency and half-life go hand-in-hand. However, while the half-life of the human IgG2 subclass is comparable to IgG1, the placental transport of IgG2 is not, with the neonatal IgG1 levels generally exceeding maternal levels at birth, but not for IgG2. We hypothesized that the unique short-hinged structure of IgG2, which enables its κ-, but not λ-isotype to form at least three different structural isoforms, might be a contributing factor to these differences. To investigate whether there was any preference for either light chain, we measured placental transport of IgG subclasses as well as κ/λ-light chain isotypes of IgG1 and IgG2 in 27 matched mother-child pairs. We also studied the half-life of IgG1 and IgG2 light chain isotypes in mice, as well as that of synthesized IgG2 structural isotypes κA and κB. In order to investigate serum clearance of IgG1 and IgG2 light-chain isotypes in humans, we quantified the relative proportions of IgG1 and IgG2 light chains in hypogammaglobulinemia patients four weeks after IVIg infusion and compared to the original IVIg isotype composition. None of our results indicate any light chain preference in either of the FcRn mediated mechanisms; half-life extension or maternal transport.
Collapse
Affiliation(s)
- Helga K. Einarsdottir
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Nigel M. Stapleton
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Sicco Scherjon
- Department of Obstetrics and Gynacology, University Medical Centre, Groningen, The Netherlands
| | - Jan Terje Andersen
- Centre for Immune Regulation (CIR) and Oslo University Hospital, Department of Immunology, Oslo, Norway
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - C. Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
32
|
Chaparro M, Gisbert JP. How safe is infliximab therapy during pregnancy and lactation in inflammatory bowel disease? Expert Opin Drug Saf 2014; 13:1749-62. [DOI: 10.1517/14740338.2014.959489] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
Development of a simple, cost-effective, semi-correlative light and electron microscopy method to allow the immunoelectron localisation of non-uniformly distributed placental proteins. Placenta 2014; 35:223-7. [DOI: 10.1016/j.placenta.2014.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/07/2014] [Accepted: 01/14/2014] [Indexed: 11/21/2022]
|
34
|
Azim Jr HA, Azim H, Peccatori FA. Treatment of cancer during pregnancy with monoclonal antibodies: a real challenge. Expert Rev Clin Immunol 2014; 6:821-6. [DOI: 10.1586/eci.10.77] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Detková D, Español T. An update on treatment strategies for common variable immunodeficiency. Expert Rev Clin Immunol 2014; 5:381-90. [DOI: 10.1586/eci.09.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Borghesi J, Mario LC, Rodrigues MN, Favaron PO, Miglino MA. Immunoglobulin Transport during Gestation in Domestic Animals and Humans—A Review. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/ojas.2014.45041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Safety of anti-TNF agents during pregnancy and breastfeeding in women with inflammatory bowel disease. Am J Gastroenterol 2013; 108:1426-38. [PMID: 23752881 DOI: 10.1038/ajg.2013.171] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/30/2013] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Anti-tumor necrosis factor (TNF) drugs are an effective therapeutic option in patients with inflammatory bowel disease (IBD). However, data regarding their safety during pregnancy and breastfeeding are scarce. The aim of this study was to critically review available data on the safety of anti-TNF therapy during pregnancy and breastfeeding in women with IBD. METHODS Bibliographical searches (MEDLINE) up to January 2013. RESULTS The studies included provided data from 462 women with IBD exposed to anti-TNF agents during pregnancy. Although these drugs cross the placenta from the end of the second trimester, they are low-risk in the short term. The use of anti-TNF agents after the second trimester leads to intra-uterine exposure. An increase in infections has recently been observed in infants exposed to immunomodulators plus anti-TNF drugs in utero, thus raising concerns about the consequences for the development of the immune system. Accordingly, it has recently been suggested that anti-TNF drugs should be stopped during the second trimester. Certolizumab is a Fab fragment of an anti-TNF monoclonal antibody, and, therefore, it may not be necessary to stop it during pregnancy. Anti-TNF drugs have been detected in breast milk, although in miniscule amounts. Case reports do not suggest toxicity; however, the effects of exposure on the neonate merit further investigation. CONCLUSIONS Anti-TNF drugs can cross the placenta from the latter part of the second trimester of gestation, although they seem to be safe, at least in the short term. Miniscule amounts of anti-TNF drugs are transferred in breast milk; therefore, a deleterious effect of this exposure on the neonate, although unlikely, cannot be excluded.
Collapse
|
38
|
Nasr A, Allam G, Al-Zahrani A, Alsulaimani A. Neonatal infections in Saudi Arabia: association with C-reactive protein, CRP -286 (C>T>A) gene polymorphism and IgG antibodies. BMC Immunol 2013; 14:38. [PMID: 23941472 PMCID: PMC3751442 DOI: 10.1186/1471-2172-14-38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 08/12/2013] [Indexed: 11/29/2022] Open
Abstract
Background C-reactive protein (CRP) is a nonspecific, acute-phase protein that rises in response to infectious and non-infectious inflammatory processes. Infections are the single largest cause of neonatal deaths globally. The primary aim of this study is to examine the association between CRP gene polymorphism and serum levels of CRP in correlation with early onset sepsis (EOS) infection in newborns living in Taif city, Saudi Arabia. The second aim is to examine the relationship between specific IgG/IgG subclasses and early onset sepsis (EOS) infection among these newborns. Methods Staphylococcus aureus (S. aureus) is one of the most common organisms related to sepsis infection in the newborn at King Abdel Aziz Specialist Hospital (KAASH). This study was conducted in Taif city, at KAASH’s neonatal intensive care unit between March and August 2012. Neonates were consecutively enrolled onto the study having met our inclusion criteria (as per our research protocol). The CRP concentration level was analysed using NycoCard® CRP Single Test. CRP -286 (C>T>A) A polymorphisms were analyzed using Pyrosequencing technology for CRP genotyping. IgG subclasses were analysed in the study population using ELISA. Result Logistic regression analyses showed that the AA and AC genotypes were negatively associated amongst EOS neonates compared to suspected neonates. The frequency of CC and CT were significantly associated with the EOS neonates compared to the suspected group. The levels of specific IgG1, IgG2 and IgG3 antibodies were significantly lower amongst EOS compared to the suspected group. Conclusions Taken together, the CRP-286 (C>T>A) A genotype polymorphism and specific IgG antibodies isotype levels can contribute to a reduced risk of EOS. Furthermore, CRP has a potential use in detecting EOS in neonates, which may mean earlier detection and management of EOS and subsequently better clinical outcome.
Collapse
Affiliation(s)
- Amre Nasr
- Department of Microbiology, College of Medicine, Taif University, Taif, SaudiArabia
| | | | | | | |
Collapse
|
39
|
A novel hypothesis for an alkaline phosphatase 'rescue' mechanism in the hepatic acute phase immune response. Biochim Biophys Acta Mol Basis Dis 2013; 1832:2044-56. [PMID: 23899605 DOI: 10.1016/j.bbadis.2013.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/10/2013] [Accepted: 07/22/2013] [Indexed: 12/24/2022]
Abstract
The liver isoform of the enzyme alkaline phosphatase (AP) has been used classically as a serum biomarker for hepatic disease states such as hepatitis, steatosis, cirrhosis, drug-induced liver injury, and hepatocellular carcinoma. Recent studies have demonstrated a more general anti-inflammatory role for AP, as it is capable of dephosphorylating potentially deleterious molecules such as nucleotide phosphates, the pathogenic endotoxin lipopolysaccharide (LPS), and the contact clotting pathway activator polyphosphate (polyP), thereby reducing inflammation and coagulopathy systemically. Yet the mechanism underlying the observed increase in liver AP levels in circulation during inflammatory insults is largely unknown. This paper hypothesizes an immunological role for AP in the liver and the potential of this system for damping generalized inflammation along with a wide range of ancillary pathologies. Based on the provided framework, a mechanism is proposed in which AP undergoes transcytosis in hepatocytes from the canalicular membrane to the sinusoidal membrane during inflammation and the enzyme's expression is upregulated as a result. Through a tightly controlled, nucleotide-stimulated negative feedback process, AP is transported in this model as an immune complex with immunoglobulin G by the asialoglycoprotein receptor through the cell and secreted into the serum, likely using the receptor's State 1 pathway. The subsequent dephosphorylation of inflammatory stimuli by AP and uptake of the circulating immune complex by endothelial cells and macrophages may lead to decreased inflammation and coagulopathy while providing an early upstream signal for the induction of a number of anti-inflammatory gene products, including AP itself.
Collapse
|
40
|
Seroprevalence and placental transmission of maternal antibodies specific for Neisseria meningitidis Serogroups A, C, Y and W135 and influence of maternal antibodies on the immune response to a primary course of MenACWY-CRM vaccine in the United Kingdom. Pediatr Infect Dis J 2013; 32:768-76. [PMID: 23538521 DOI: 10.1097/inf.0b013e318292f425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Maternal antibodies give neonates some protection against bacterial infection. We measured antibodies against Neisseria meningitidis serogroups A, C, Y and W135 in mothers and their 2-month-old infants at study enrollment. We also assessed the impact of maternal antibody present at 2 months of age on the immune response to a primary course of quadrivalent meningococcal conjugate vaccine (MenACWY-CRM197) given at 2 and 4 months of age. METHODS This was a single-center, open-label, randomized study undertaken in Oxford, United Kingdom. Two hundred sixteen healthy infants were enrolled in the study and vaccinated with MenACWY-CRM197 at 2 and 4 months of age. Blood was obtained from all mothers, in a subset of infants at 2 months and all infants at 5 months. Antibody and memory B-cell responses at 5 months were correlated with maternal antibodies. RESULTS Mothers had low IgG antibodies against serogroups C, W135 and Y polysaccharides, but high serogroup A antibody, whereas 61-78% had protective human complement serum bactericidal activity (hSBA) (≥1:4) for serogroups C, W135 and Y but only 31% for serogroup A. Only 9%, 32%, 45% and 19% of 2-month-old infants had hSBA ≥1:4 for serogroups A, C, W135 and Y, respectively. Maternal antibody had little association on responses to MenACWY-CRM197, except a moderate negative association between MenC-specific bactericidal antibody at 2 and 5 months (r = -0.5, P = 0.006, n = 28) and between carrier-specific IgG antibody at 2 months and MenC-specific hSBA/IgG antibody at 5 months (r = -0.4, P = 0.02 and 0.04, n = 32 and 23). Nonetheless, 90% of infants achieved protective MenC-hSBA titers after vaccination at 2 and 4 months of age. CONCLUSIONS The levels of serogroup-specific meningococcal antibodies were low in mothers and 2-month-old infants. Immunizing mothers before or during pregnancy with meningococcal conjugate vaccines might increase antibody levels in early infancy and provide protection against infection due to N. meningitidis.
Collapse
|
41
|
Sakuma Y, Baba R, Arita K, Morimoto H, Fujita M. Food allergens are transferred intact across the rat blood-placental barrier in vivo. Med Mol Morphol 2013; 47:14-20. [PMID: 23475277 DOI: 10.1007/s00795-013-0029-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 12/07/2012] [Indexed: 10/27/2022]
Abstract
We investigated the mechanism of transplacental macromolecular transport in rats on the nineteenth day of pregnancy using tracers, transmission electron microscopy and immunohistochemistry. The blood-placental barrier of full-term rat placentas was composed of a trilaminar layer of trophoblast cells that separates the fetal capillaries from the maternal blood spaces: a layer of cytotrophoblasts lining the maternal blood space and a bilayer of syncytiotrophoblast surrounding the fetal capillaries. Horseradish peroxidase, intravenously injected into the maternal circulation, was found in the maternal blood spaces, the interspaces between the cytotrophoblasts and the syncytiotrophoblast I, many pits and small vesicles in the syncytiotrophoblast I, vesicles of the syncytiotrophoblast II, fetal connective tissue and fetal capillaries. Intravenously injected ovalbumin was detected in the maternal blood spaces, a trilaminar layer and the fetal capillaries. Neonatal Fc receptor (FcRn), a receptor for IgG, was localized at the maternal side of the blood-placental barrier. These results show that the structure of the rat blood-placental barrier is quite similar to the human blood-placental barrier, and non-specific macromolecules and food allergens may penetrate through the blood-placental barrier of the full-term placenta from the maternal to fetal circulation mediated by FcRn.
Collapse
Affiliation(s)
- Yoshiko Sakuma
- Graduate School of Health and Nutrition Sciences, Nakamura Gakuen University, 5-7-1 Befu, Jyonan-ku, Fukuoka, 814-0198, Japan
| | | | | | | | | |
Collapse
|
42
|
Takizawa T, Robinson JM. Correlative fluorescence and transmission electron microscopy in tissues. Methods Cell Biol 2012; 111:37-57. [PMID: 22857922 DOI: 10.1016/b978-0-12-416026-2.00003-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Correlative microscopy has meant different things over the years; currently, this term refers to imaging the same exact structures with two or more imaging modalities. This commonly involves combining fluorescence and electron microscopy. Much of the recent work related to correlative microscopy has been done using cell culture models. However, many biological questions cannot be addressed in these models, but require instead the 3-dimensional organization of cells found in tissues. Herein, we discuss some of the issues related to correlative microscopy of tissues including the major reporter systems presently available for correlative microscopy. We present data from our own work in which we have focused on the use of ultrathin cryosections of tissues as the substrate for immunolabeling to combine immunofluorescence and electron microscopy of the same sub-cellular structures.
Collapse
Affiliation(s)
- Toshihiro Takizawa
- Department of Molecular Anatomy, Nippon Medical School, Tokyo 113-8602, Japan
| | | |
Collapse
|
43
|
Ellinger I, Fuchs R. HFcRn-mediated transplacental immunoglobulin G transport: protection of and threat to the human fetus and newborn. Wien Med Wochenschr 2012; 162:207-13. [PMID: 22717875 DOI: 10.1007/s10354-012-0085-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 02/28/2012] [Indexed: 02/07/2023]
Abstract
In human newborns, endogenous levels of plasma immunoglobulin G (IgG) begin to rise slowly after birth following exposure to the environment. For immunoprotection during fetal and early neonatal life, maternal IgG is provided by transplacental transport. While cellular immunoprotective IgG effects are mainly triggered by FcγRI, -RII and -RIII, transplacental IgG transfer is mediated by the MHC class I-like neonatal Fc-receptor, hFcRn. This compact review explains the mechanism of hFcRn-mediated IgG transcytosis across the placental barrier - syncytiotrophoblast and fetal endothelial cells. Restrictions of this IgG transport are summarized. These include IgG subclass discrimination and limited IgG transport before the third trimester that can cause insufficient protection from infections of preterm (≤ 35 th week) delivered babies. As hFcRn does not discriminate beneficial from hazardous IgGs, maternal auto- and alloimmune as well as therapeutic antibodies can reach the fetus. The consequences including severe diseases of the newborn are summarized in this article.
Collapse
Affiliation(s)
- Isabella Ellinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria.
| | | |
Collapse
|
44
|
Ganesan LP, Kim J, Wu Y, Mohanty S, Phillips GS, Birmingham DJ, Robinson JM, Anderson CL. FcγRIIb on liver sinusoidal endothelium clears small immune complexes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:4981-8. [PMID: 23053513 PMCID: PMC4381350 DOI: 10.4049/jimmunol.1202017] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
It has long been known that the ITIM-bearing IgG Fc receptor (FcγRIIb, RIIb) is expressed on liver sinusoidal endothelial cells (LSEC) and that the liver is the major site of small immune complex (SIC) clearance. Thus, we proposed that RIIb of LSEC eliminates blood-borne SIC, thereby controlling immune complex-mediated autoimmune disease. Testing this hypothesis, we found most RIIb of the mouse, fully three-quarters, to be expressed in liver. Moreover, most (90%) liver RIIb was expressed in LSEC, the remainder in Kupffer cells. An absent FcRγ in LSEC implied that RIIb is the sole FcγR expressed. Testing the capacity of liver RIIb to clear blood-borne SIC, we infused mice intravenously with radio-iodinated SIC made of OVA and rabbit IgG anti-OVA. Tracking decay of SIC from the blood, we found the RIIb knockout strain to be severely deficient in eliminating SIC compared with the wild-type strain, terminal half-lives being 6 and 1.5 h, respectively. RIIb on LSEC, a major scavenger, keeps SIC blood concentrations low and minimizes pathologic deposition of inflammatory immune complex.
Collapse
Affiliation(s)
- Latha P. Ganesan
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jonghan Kim
- Harvard School of Public Health, Boston, MA, 02115, USA
| | - Yun Wu
- NSF Nanoscale Science and Engineering Center for Nanoengineering of Polymer Biomedical Devices, The Ohio State University, Columbus, OH, 43210, USA
| | - Sudhasri Mohanty
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Gary S. Phillips
- Department of Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Daniel J. Birmingham
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - John M. Robinson
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Clark L. Anderson
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
45
|
Walton JR, Frey HA, Vandre DD, Kwiek JJ, Ishikawa T, Takizawa T, Robinson JM, Ackerman WE. Expression of flotillins in the human placenta: potential implications for placental transcytosis. Histochem Cell Biol 2012; 139:487-500. [PMID: 23064789 DOI: 10.1007/s00418-012-1040-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2012] [Indexed: 02/07/2023]
Abstract
A proteomics survey of human placental syncytiotrophoblast (ST) apical plasma membranes revealed peptides corresponding to flotillin-1 (FLOT1) and flotillin-2 (FLOT2). The flotillins belong to a class of lipid microdomain-associated integral membrane proteins that have been implicated in clathrin- and caveolar-independent endocytosis. In the present study, we characterized the expression of the flotillin proteins within the human placenta. FLOT1 and FLOT2 were coexpressed in placental lysates and BeWo human trophoblast cells. Immunofluorescence microscopy of first-trimester and term placentas revealed that both proteins were more prominent in villous endothelial cells and cytotrophoblasts (CTs) than the ST. Correspondingly, forskolin-induced fusion in BeWo cells resulted in a decrease in FLOT1 and FLOT2, suggesting that flotillin protein expression is reduced following trophoblast syncytialization. The flotillin proteins co-localized with a marker of fluid-phase pinocytosis, and knockdown of FLOT1 and/or FLOT2 expression resulted in decreased endocytosis of cholera toxin B subunit. We conclude that FLOT1 and FLOT2 are abundantly coexpressed in term villous placental CTs and endothelial cells, and in comparison, expression of these proteins in the ST is reduced. These findings suggest that flotillin-dependent endocytosis is unlikely to be a major pathway in the ST, but may be important in the CT and endothelium.
Collapse
Affiliation(s)
- Janelle R Walton
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
DeSesso JM, Williams AL, Ahuja A, Bowman CJ, Hurtt ME. The placenta, transfer of immunoglobulins, and safety assessment of biopharmaceuticals in pregnancy. Crit Rev Toxicol 2012; 42:185-210. [PMID: 22348352 DOI: 10.3109/10408444.2011.653487] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
47
|
Hůlková H, Ledvinová J, Kuchař L, Šmíd F, Honzíková J, Elleder M. Glycosphingolipid profile of the apical pole of human placental capillaries: the relevancy of the observed data to Fabry disease. Glycobiology 2012; 22:725-32. [PMID: 22319058 DOI: 10.1093/glycob/cws050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A series of six full-term placentas and umbilical cords were examined using the in situ detection of globotriaosylceramide (Gb3Cer), GM1 ganglioside (GM1), GM3 ganglioside (GM3), cholesterol and caveolin 1. Immunohistochemical study showed uniform distinct staining of the apical membrane of villous capillary endothelial cells for Gb3Cer, GM1, GM3 and cholesterol. There was also a strong signal for caveolin 1. The immunophenotype suggests the presence of caveola-associated raft microdomains. The immunophenotype was almost completely shared with the extravillous intravascular trophoblast in the basal plate. It was absent in the endothelial cells of umbilical vessels and in the capillaries of somatic structures (heart, lung, skeletal muscle and skin) in neonates as well as in adults, including capillaries of the proliferative endometrium. Results of in situ analyses were confirmed by lipid chromatographic analysis of tissue homogenates and by tandem mass spectrometry. Lysosomal Gb3Cer turnover was followed in three placentas including umbilical cords from Fabry disease (α-galactosidase A deficiency). Lysosomal storage was restricted to vascular smooth muscle cells and to endothelial cells of umbilical vessels. Placental villous capillary endothelial cells displaying a strong non-lysosomal staining for Gb3Cer were free of lysosomal storage.
Collapse
Affiliation(s)
- Helena Hůlková
- Institute of Inherited Metabolic Disorders, First Faculty of Medicine and General University Hospital, Charles University in Prague, Ke Karlovu 2, 120 00 Praha 2, Czech Republic
| | | | | | | | | | | |
Collapse
|
48
|
The expression and function of fatty acid transport protein-2 and -4 in the murine placenta. PLoS One 2011; 6:e25865. [PMID: 22028793 PMCID: PMC3197585 DOI: 10.1371/journal.pone.0025865] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/13/2011] [Indexed: 11/19/2022] Open
Abstract
Background The uptake and trans-placental trafficking of fatty acids from the maternal blood into the fetal circulation are essential for embryonic development, and involve several families of proteins. Fatty acid transport proteins (FATPs) uniquely transport fatty acids into cells. We surmised that placental FATPs are germane for fetal growth, and are regulated during hypoxic stress, which is associated with reduced fat supply to the fetus. Methodology/Principal Findings Using cultured primary term human trophoblasts we found that FATP2, FATP4 and FATP6 were highly expressed in trophoblasts. Hypoxia enhanced the expression of trophoblastic FATP2 and reduced the expression of FATP4, with no change in FATP6. We also found that Fatp2 and Fatp4 are expressed in the mouse amnion and placenta, respectively. Mice deficient in Fatp2 or Fatp4 did not deviate from normal Mendelian distribution, with both embryos and placentas exhibiting normal weight and morphology, triglyceride content, and expression of genes related to fatty acid mobilization. Conclusions/Significance We conclude that even though hypoxia regulates the expression of FATP2 and FATP4 in human trophoblasts, mouse Fatp2 and Fatp4 are not essential for intrauterine fetal growth.
Collapse
|
49
|
Abstract
A growing body of evidence highlights the importance of a mother's nutrition from preconception through lactation in programming the emerging organ systems and homeostatic pathways of her offspring. The developing immune system may be particularly vulnerable. Indeed, examples of nutrition-mediated immune programming can be found in the literature on intra-uterine growth retardation, maternal micronutrient deficiencies, and infant feeding. Current models of immune ontogeny depict a "layered" expansion of increasingly complex defenses, which may be permanently altered by maternal malnutrition. One programming mechanism involves activation of the maternal hypothalamic-pituitary-adrenal axis in response to nutritional stress. Fetal or neonatal exposure to elevated stress hormones is linked in animal studies to permanent changes in neuroendocrine-immune interactions, with diverse manifestations such as an attenuated inflammatory response or reduced resistance to tumor colonization. Maternal malnutrition may also have a direct influence, as evidenced by nutrient-driven epigenetic changes to developing T regulatory cells and subsequent risk of allergy or asthma. A 3rd programming pathway involves placental or breast milk transfer of maternal immune factors with immunomodulatory functions (e.g. cytokines). Maternal malnutrition can directly affect transfer mechanisms or influence the quality or quantity of transferred factors. The public health implications of nutrition-mediated immune programming are of particular importance in the developing world, where prevalent maternal undernutrition is coupled with persistent infectious challenges. However, early alterations to the immune system, resulting from either nutritional deficiencies or excesses, have broad relevance for immune-mediated diseases, such as asthma, and chronic inflammatory conditions like cardiovascular disease.
Collapse
|
50
|
Goto Y, Lee YA. Is schizophrenia developmental adaptation to environmental menaces? Med Hypotheses 2011; 77:756-62. [PMID: 21840133 DOI: 10.1016/j.mehy.2011.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 06/16/2011] [Accepted: 07/18/2011] [Indexed: 11/19/2022]
Abstract
Schizophrenia is a devastating mental disorder, with its symptoms typically emerging during late adolescence to young adulthood. In contrast, accumulating evidence suggests that schizophrenia is a developmental disorder in which brain abnormalities may occur even before birth. This has brought the major challenge to explain such discrepancy of brain deficits occurring during prenatal period and emergence of symptoms during adulthood. A number of ideas have been proposed to explain delayed emergence of symptoms at adulthood in relation to maturational processes of various brain systems during adolescence. However, these still lack clear relationship to prenatal deficits. Thus, a key to better understand the pathology of schizophrenia is to unveil a theory or model that can explain the relationship between prenatal deficits and post-pubertal onset of symptoms. Here we propose a novel hypothesis, along with discussion of several lines of evidences supporting it, that schizophrenia may not be a disorder in a strict sense, but rather be understood as the biological state occurring as consequence of adaptation to severe environmental conditions during the prenatal periods, which explains the relationship between prenatal developmental deficits and the postnatal maturational process for onset of symptoms.
Collapse
Affiliation(s)
- Yukiori Goto
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | | |
Collapse
|