51
|
Chen YH, Liu ZB, Ma L, Zhang ZC, Fu L, Yu Z, Chen W, Song YP, Wang P, Wang H, Xu DX. Gestational vitamin D deficiency causes placental insufficiency and fetal intrauterine growth restriction partially through inducing placental inflammation. J Steroid Biochem Mol Biol 2020; 203:105733. [PMID: 32784046 DOI: 10.1016/j.jsbmb.2020.105733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/24/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Several epidemiological studies suggest an association between vitamin D deficiency (VDD) and fetal intrauterine growth restriction (IUGR). Here, we explored the mechanism through which VDD induced fetal IUGR. Pregnant mice were fed with VDD diet to establish VDD model. Cyp27b1+/- mice were generated to develop a model of active vitamin D3 deficiency. Cyp27b1+/- mice were injected with either 1α,25(OH)2D3 or vehicle once a day throughout pregnancy. As expected, fetal weight and crown-rump length were reduced in VDD diet-fed mice. Correspondingly, fetal weight and crown-rump length were lower in cyp27b1+/- mice. 1α,25(OH)2D3 elevated fetal weight and crown-rump length, and protected cyp27b1+/- mice from fetal IUGR. Further analysis found that placental proliferation was inhibited and placental weight was decreased in VDD diet-fed mice. Several growth factors and nutrient transfer pumps were downregulated in the placentas of VDD diet-fed mice. Mechanistically, several inflammatory cytokines were upregulated and placental NF-κB was activated not only in VDD diet-fed mice but also in VDD pregnant women. Interestingly, 1α,25(OH)2D3 inhibited the downregulated of placental nutrient transfer pumps and the upregulated of placental inflammatory cytokines in Cyp27b1+/- mice. These results provide experimental evidence that gestational VDD causes placental insufficiency and fetal IUGR may be through inducing placental inflammation.
Collapse
Affiliation(s)
- Yuan Hua Chen
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China; Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Zhi Bing Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Li Ma
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Zhi Cheng Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Lin Fu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Zhen Yu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Wei Chen
- Department of Histology and Embryology, Anhui Medical University, Hefei, 230032, China
| | - Ya Ping Song
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Peng Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - De Xiang Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Department of Toxicology, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
52
|
Abstract
Vertical transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and possible induction of pregnancy complications, including miscarriage, fetal malformations, fetal growth restriction and/or stillbirth, are serious concerns for pregnant individuals with COVID-19. According to clinical information, the incidence of vertical transmission of SARS-CoV-2 is limited to date. However, even if a neonate tests negative for SARS-CoV-2, frequent abnormal findings, including fetal and maternal vascular malperfusion, have been reported in cases of COVID-19-positive mothers. Primary receptor of SARS-CoV-2 is estimated as angiotensin-converting enzyme 2 (ACE2). It is highly expressed in maternal-fetal interface cells, such as syncytiotrophoblasts, cytotrophoblasts, endothelial cells, and the vascular smooth muscle cells of primary and secondary villi. However other route of transplacental infection cannot be ruled out. Pathological examinations have demonstrated that syncytiotrophoblasts are often infected with SARS-CoV-2, but fetuses are not always infected. These findings suggest the presence of a placental barrier, even if it is not completely effective. As the frequency and molecular mechanisms of intrauterine vertical transmission of SARS-CoV-2 have not been determined to date, intensive clinical examinations by repeated ultrasound and fetal heart rate monitoring are strongly recommended for pregnant women infected with COVID-19. In addition, careful investigation of placental samples after delivery by both morphological and molecular methods is also strongly recommended.
Collapse
Affiliation(s)
- Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
53
|
Abstract
Platelets, small anucleate cells circulating in the blood, are critical mediators in haemostasis and thrombosis. Interestingly, recent studies demonstrated that platelets contain both pro-inflammatory and anti-inflammatory molecules, equipping platelets with immunoregulatory function in both innate and adaptive immunity. In the context of infectious diseases, platelets are involved in early detection of invading microorganisms and are actively recruited to sites of infection. Platelets exert their effects on microbial pathogens either by direct binding to eliminate or restrict dissemination, or by shaping the subsequent host immune response. Reciprocally, many invading microbial pathogens can directly or indirectly target host platelets, altering platelet count or/and function. In addition, microbial pathogens can impact the host auto- and alloimmune responses to platelet antigens in several immune-mediated diseases, such as immune thrombocytopenia, and fetal and neonatal alloimmune thrombocytopenia. In this review, we discuss the mechanisms that contribute to the bidirectional interactions between platelets and various microbial pathogens, and how these interactions hold relevant implications in the pathogenesis of many infectious diseases. The knowledge obtained from "well-studied" microbes may also help us understand the pathogenesis of emerging microbes, such as SARS-CoV-2 coronavirus.
Collapse
Affiliation(s)
- Conglei Li
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
| | - June Li
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
| | - Heyu Ni
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
54
|
Freitag N, Tirado-Gonzalez I, Barrientos G, Powell KL, Boehm-Sturm P, Koch SP, Hecher K, Staff AC, Arck PC, Diemert A, Blois SM. Galectin-3 deficiency in pregnancy increases the risk of fetal growth restriction (FGR) via placental insufficiency. Cell Death Dis 2020; 11:560. [PMID: 32703931 PMCID: PMC7378206 DOI: 10.1038/s41419-020-02791-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 12/26/2022]
Abstract
Fetal growth restriction (FGR) is the most common pregnancy complication in developed countries. Pregnancies affected by FGR, frequently concur with complications and high risk of neonatal morbidity and mortality. To date, no approved treatment is available for pregnant women affected with FGR. The objective of this study was to investigate the contribution of galectin-3 (gal-3), a β-galactoside binding protein involved in pregnancy, placental function and fetal growth. We demonstrated that lack of gal-3 during mouse pregnancy leads to placental dysfunction and drives FGR in the absence of a maternal preeclampsia syndrome. Analysis of gal-3 deficient dams revealed placental inflammation and malperfusion, as well as uterine natural killer cell infiltration with aberrant activation. Our results also show that FGR is associated with a failure to increase maternal circulating gal-3 levels during the second and third trimester in human pregnancies. Placentas from human pregnancies affected by FGR displayed lower gal-3 expression, which correlated with placental dysfunction. These data highlight the importance of gal-3 in the promotion of proper placental function, as its absence leads to placental disease and subsequent FGR.
Collapse
Affiliation(s)
- Nancy Freitag
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and Charité - Universitätsmedizin Berlin, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for Psychosomatic Medicine, Berlin, Germany
| | - Irene Tirado-Gonzalez
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Katie L Powell
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW, 2065, Australia
| | - Philipp Boehm-Sturm
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin, and Berlin Institute of Health, Department of Experimental Neurology, Center for Stroke Research, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan P Koch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin, and Berlin Institute of Health, Department of Experimental Neurology, Center for Stroke Research, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, 20251, Germany
| | - Anne C Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Division of Obstetrics and Gyneacology, Oslo University Hospital, Oslo, Norway
| | - Petra C Arck
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, 20251, Germany
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, 20251, Germany
| | - Sandra M Blois
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association, and Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, 20251, Germany.
| |
Collapse
|
55
|
St-Germain LE, Castellana B, Baltayeva J, Beristain AG. Maternal Obesity and the Uterine Immune Cell Landscape: The Shaping Role of Inflammation. Int J Mol Sci 2020; 21:E3776. [PMID: 32471078 PMCID: PMC7312391 DOI: 10.3390/ijms21113776] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is often equated to the physiological response to injury or infection. Inflammatory responses defined by cytokine storms control cellular mechanisms that can either resolve quickly (i.e., acute inflammation) or remain prolonged and unabated (i.e., chronic inflammation). Perhaps less well-appreciated is the importance of inflammatory processes central to healthy pregnancy, including implantation, early stages of placentation, and parturition. Pregnancy juxtaposed with disease can lead to the perpetuation of aberrant inflammation that likely contributes to or potentiates maternal morbidity and poor fetal outcome. Maternal obesity, a prevalent condition within women of reproductive age, associates with increased risk of developing multiple pregnancy disorders. Importantly, chronic low-grade inflammation is thought to underlie the development of obesity-related obstetric and perinatal complications. While diverse subsets of uterine immune cells play central roles in initiating and maintaining healthy pregnancy, uterine leukocyte dysfunction as a result of maternal obesity may underpin the development of pregnancy disorders. In this review we discuss the current knowledge related to the impact of maternal obesity and obesity-associated inflammation on uterine immune cell function, utero-placental establishment, and pregnancy health.
Collapse
Affiliation(s)
- Lauren E. St-Germain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Barbara Castellana
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Jennet Baltayeva
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Alexander G. Beristain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| |
Collapse
|
56
|
Liu Y, Wu Y, Tian M, Luo W, Zhang C, Liu Y, Li K, Cheng W, Liu D. Protein Expression Profile in IVF Follicular Fluid and Pregnancy Outcome Analysis in Euthyroid Women with Thyroid Autoimmunity. ACS OMEGA 2020; 5:11439-11447. [PMID: 32478232 PMCID: PMC7254522 DOI: 10.1021/acsomega.0c00463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/30/2020] [Indexed: 05/05/2023]
Abstract
The objective of this study is to investigate the influence of the thyroid autoantibodies on the protein expression in follicular fluid and the clinical outcome of assisted reproductive technology. A total of 602 patients treated for infertility were screened; 49 euthyroid women who were positive for thyroid autoantibodies and 63 negative controls were recruited. Follicular fluid samples were analyzed using proteomics. Validation of target proteins in follicular fluid was performed by using parallel reaction monitoring. Differentially expressed proteins in follicular fluid, clinical pregnancy rate, abortion rate, and live-birth rate were analyzed. Clinical pregnancy rates and take-home baby rates in the thyroid autoimmunity (TAI) group were less than in the control group, but abortion rates in the TAI group were higher than in the control group (all P < 0.005). A total of 49 proteins were differentially expressed in the TAI-positive group. In Gene Ontology secondary annotations of all the proteins identified, five types of proteins were associated with the reproductive process. Among 11 proteins quantitatively identified by parallel reaction monitoring, angiotensinogen and fetuin-B were associated with reproduction. These differentially expressed proteins identified in this study involved multiple pathways according to the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. Our study provides evidence that some differentially expressed proteins between TAI-positive women and controls were associated with the reproductive process and closely related to important physiologic effects, which could partially explain the underlying mechanism link between TAI and the adverse outcomes of assisted reproductive technology.
Collapse
Affiliation(s)
- Yuting Liu
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Department
of Endocrinology and Metabolism, The Ninth
People’s Hospital of Chongqing, Chongqing 400700, China
| | - Yijia Wu
- Reproductive
Medical Center, Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical
University, Chongqing 400010, China
| | - Mingyuan Tian
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Wenwen Luo
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chanyu Zhang
- Reproductive
Medical Center, Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical
University, Chongqing 400010, China
| | - Yongjian Liu
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ke Li
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Wei Cheng
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Dongfang Liu
- Department
of Endocrinology and Metabolism, The Second
Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- ,
| |
Collapse
|
57
|
Abstract
The disease course of autoimmune diseases such as rheumatoid arthritis is altered during pregnancy, and a similar modulatory role of pregnancy on inflammatory bowel disease (IBD) has been proposed. Hormonal, immunological, and microbial changes occurring during normal pregnancy may interact with the pathophysiology of IBD. IBD consists of Crohn's disease and ulcerative colitis, and because of genetic, immunological, and microbial differences between these disease entities, they may react differently during pregnancy and should be described separately. This review will address the pregnancy-induced physiological changes and their potential effect on the disease course of ulcerative colitis and Crohn's disease, with emphasis on the modulation of epithelial barrier function and immune profiles by pregnancy hormones, microbial changes, and microchimerism.
Collapse
|
58
|
Quinn KE, Matson BC, Caron KM. Deletion of atypical chemokine receptor 3 (ACKR3) increases immune cells at the fetal-maternal interface. Placenta 2020; 95:18-25. [PMID: 32452398 DOI: 10.1016/j.placenta.2020.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/16/2020] [Accepted: 04/17/2020] [Indexed: 11/18/2022]
Abstract
Establishment of immune cell populations and adaptations in immune cells are critical aspects during pregnancy that lead to protection of the semi-allogenic fetus. Appropriate immune cell activation and trophoblast migration are regulated in part by chemokines, the availability of which can be fine-tuned by decoy receptors. Atypical chemokine receptor 3 (ACKR3), previously named C-X-C chemokine receptor 7 (CXCR7), is a chemokine decoy receptor expressed in placenta, but little is known about how this receptor affects placental development. In this study, we investigated the phenotypic characteristics of placentas from Ackr3-/- embryos to determine how Ackr3 contributes to early placentation. In placentas from Ackr3-/- embryos, we observed an increase in decidual compaction and in the size of the uterine natural killer cell population. Ackr3 knockdown in trophoblast cells led to a decrease in trophoblast migration. These findings suggest that this decoy receptor may therefore be an important factor in normal placentation.
Collapse
Affiliation(s)
- Kelsey E Quinn
- Department of Cell Biology and Physiology, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB# 7545, Chapel Hill, NC, 27599, USA.
| | - Brooke C Matson
- Department of Cell Biology and Physiology, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB# 7545, Chapel Hill, NC, 27599, USA.
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB# 7545, Chapel Hill, NC, 27599, USA; Department of Genetics, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB# 7545, Chapel Hill, NC, 27599, USA; Lineberger Comprehensive Cancer Center, 111 Mason Farm Road, 6312B Medical Biomolecular Research Building, CB# 7545, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
59
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
60
|
Molecular characteristics and possible functions of innate lymphoid cells in the uterus and gut. Cytokine Growth Factor Rev 2020; 52:15-24. [DOI: 10.1016/j.cytogfr.2019.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/23/2022]
|
61
|
Hamsanathan S, Alder JK, Sellares J, Rojas M, Gurkar AU, Mora AL. Cellular Senescence: The Trojan Horse in Chronic Lung Diseases. Am J Respir Cell Mol Biol 2020; 61:21-30. [PMID: 30965013 DOI: 10.1165/rcmb.2018-0410tr] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Senescence is a cell fate decision characterized by irreversible arrest of proliferation accompanied by a senescence-associated secretory phenotype. Traditionally, cellular senescence has been recognized as a beneficial physiological mechanism during development and wound healing and in tumor suppression. However, in recent years, evidence of negative consequences of cellular senescence has emerged, illuminating its role in several chronic pathologies. In this context, senescent cells persist or accumulate and have detrimental consequences. In this review, we discuss the possibility that in chronic obstructive pulmonary disease, persistent senescence impairs wound healing in the lung caused by secretion of proinflammatory senescence-associated secretory phenotype factors and exhaustion of progenitor cells. In contrast, in idiopathic pulmonary fibrosis, chronic senescence in alveolar epithelial cells exacerbates the accumulation of senescent fibroblasts together with production of extracellular matrix. We review how cellular senescence may contribute to lung disease pathology.
Collapse
Affiliation(s)
| | - Jonathan K Alder
- 2 Division of Pulmonary Allergy and Critical Care Medicine, and.,3 Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases
| | - Jacobo Sellares
- 4 Interstitial Lung Disease Program, Servei de Pneumologia, Hospital Clínic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,5 Centro de Investigaciones Biomedicas en Red-Enfermedades Respiratorias (CibeRes CB06/06/0028), Instituto de Salud Carlos III, Barcelona, Spain; and
| | - Mauricio Rojas
- 2 Division of Pulmonary Allergy and Critical Care Medicine, and.,3 Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases.,6 McGowan Institute of Regenerative Medicine, and
| | - Aditi U Gurkar
- 1 Aging Institute.,7 Division of Geriatric Medicine, Department of Medicine.,8 Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Ana L Mora
- 1 Aging Institute.,2 Division of Pulmonary Allergy and Critical Care Medicine, and.,9 Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
62
|
Ya F, Xu XR, Tian Z, Gallant RC, Song F, Shi Y, Wu Y, Wan J, Zhao Y, Adili R, Ling W, Ni H, Yang Y. Coenzyme Q10 attenuates platelet integrin αIIbβ3 signaling and platelet hyper-reactivity in ApoE-deficient mice. Food Funct 2020; 11:139-152. [DOI: 10.1039/c9fo01686d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
CoQ10 supplementation in ApoE−/− mice attenuates high-fat diet-induced platelet hyper-reactivity via down-regulating platelet αIIbβ3 signaling, and thus protecting against atherothrombosis.
Collapse
|
63
|
Daimon A, Morihara H, Tomoda K, Morita N, Koishi Y, Kanki K, Ohmichi M, Asahi M. Intravenously Injected Pluripotent Stem Cell-derived Cells Form Fetomaternal Vasculature and Prevent Miscarriage in Mouse. Cell Transplant 2020; 29:963689720970456. [PMID: 33349053 PMCID: PMC7873769 DOI: 10.1177/0963689720970456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/26/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Miscarriage is the most common complication of pregnancy, and about 1% of pregnant women suffer a recurrence. Using a widely used mouse miscarriage model, we previously showed that intravenous injection of bone marrow (BM)-derived endothelial progenitor cells (EPCs) may prevent miscarriage. However, preparing enough BM-derived EPCs to treat a patient might be problematic. Here, we demonstrated the generation of mouse pluripotent stem cells (PSCs), propagation of sufficient PSC-derived cells with endothelial potential (PSC-EPs), and intravenous injection of the PSC-EPs into the mouse miscarriage model. We found that the injection prevented miscarriage. Three-dimensional reconstruction images of the decidua after tissue cleaning revealed robust fetomaternal neovascularization induced by the PSC-EP injection. Additionally, the injected PSC-EPs directly formed spiral arteries. These findings suggest that intravenous injection of PSC-EPs could become a promising remedy for recurrent miscarriage.
Collapse
Affiliation(s)
- Atsushi Daimon
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- These authors contributed equally to
this article
| | - Hirofumi Morihara
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- These authors contributed equally to
this article
| | - Kiichiro Tomoda
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
- Department of Life Science Frontiers, Center for iPS Cell Research
and Application, Kyoto University, Japan
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA,
USA
- These authors contributed equally to
this article
| | - Natsuko Morita
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
| | - Yoshinori Koishi
- Division of Research Animal Laboratory and Translational Medicine,
Research and Development Center, Takatsuki, Osaka, Japan
| | - Kazuyoshi Kanki
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College,
Takatsuki, Japan
| | - Michio Asahi
- Department of Pharmacology, Osaka Medical College, Takatsuki,
Japan
| |
Collapse
|
64
|
Abstract
Maternal alloimmunization to paternally inherited antigens on fetal/neonatal platelets can cause fetal/neonatal alloimmune thrombocytopenia (FNAIT) after antibody-mediated removal of platelets from the fetal circulation. The complications vary from mild bleeding symptoms to severe intracranial hemorrhage and subsequent neurological impairment or death. Studies on in vivo mechanisms are challenging to measure directly in pregnant women, rendering murine models as valuable and attractive alternatives, despite some critical differences between mice and men affecting the translational value. Here we present and discuss, the different murine models that substantially have increased our knowledge and understanding of FNAIT pathogenesis - as well as pre-clinical evaluation of therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Trude Victoria Rasmussen
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway; Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maria Therese Ahlen
- Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
65
|
Wang J, Liu C, Que W, Fujino M, Tong G, Yan H, Li XK. Immunomodulatory effects of Salvianolic acid B in a spontaneous abortion mouse model. J Reprod Immunol 2019; 137:103075. [PMID: 31918160 DOI: 10.1016/j.jri.2019.103075] [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: 04/27/2019] [Revised: 11/20/2019] [Accepted: 12/19/2019] [Indexed: 11/30/2022]
Abstract
Pregnancy is a kind of natural immune tolerance. Immune factors play an important role in recurrent spontaneous abortion and repeated implantation failure. Salvianolic acid B (SalB) has anti-tumor, anti-inflammatory, anti-oxidation and immunomodulatory functions. However, there are few reports on the relationship between SalB and maternal-fetal immune tolerance. In this study, CBA/J × DBA/2 J mice as a spontaneous abortion mouse model were given SalB. The results showed that the abortion rate was significantly decreased after SalB treatment. The populations of Nkp46 and cytotoxic CD8+ T cells in the placenta of female mice treated with SalB were significantly decreased. The qRT-PCR showed that SalB was able to significantly reduce the expression of pro-inflammatory factors and Toll-like Receptor in the placenta. In addition, SalB was able to increase the area of the labyrinth in the placenta. In conclusion, these findings suggest that SalB is beneficial for the immune-modulation at the maternal-fetal interface in a spontaneous abortion mouse model, resulting in a decrease in the abortion rate. This may encourage new ideas for the treatment of patients with repeated implantation failure.
Collapse
Affiliation(s)
- Jing Wang
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Chi Liu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weitao Que
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Guoqing Tong
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
66
|
Zhu Y, Liu Q, Liao M, Diao L, Wu T, Liao W, Wang Z, Li B, Zhang S, Wang S, Xie W, Jiang Y, Xu N, Zeng Y, Yang BB, Zhang Y. Overexpression of lncRNA EPB41L4A-AS1 Induces Metabolic Reprogramming in Trophoblast Cells and Placenta Tissue of Miscarriage. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:518-532. [PMID: 31671345 PMCID: PMC6838551 DOI: 10.1016/j.omtn.2019.09.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/26/2019] [Accepted: 09/14/2019] [Indexed: 11/20/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been shown to be crucial regulators in numerous human diseases. However, little is known about their effects on early recurrent miscarriage (RM). Here we aimed to investigate the role of lncRNA EPB41L4A-AS1 on placental trophoblast cell metabolic reprogramming, which might be involved in the pathogenesis of RM. After microarray and GEO database analyses, we found that EPB41L4A-AS1 was significantly increased in early RM placental tissue, and this increase may relate to estradiol-mediated upregulation of PGC-1α. EPB41L4A-AS1 overexpression inhibits glycolysis but increases the dependence on fatty acid oxidation in mitochondrion metabolism and suppresses the Warburg effect, which is necessary for rapid growth of the placental villus, leading to miscarriage. Mechanistic analyses demonstrated that EPB41L4A-AS1 functions as a lncRNA in the regulation of VDAC1 and HIF-1α expression through enhancement of H3K4me3 levels in the promoters of VDAC1 and HIF1A-AS1, a natural antisense transcript (NAT) lncRNA of HIF-1α. Taken together, these findings demonstrate that aberrant expression of EPB41L4A-AS1 is involved in the etiology of early RM, and it may be a candidate diagnostic hallmark and a potential therapeutic target for early RM treatment.
Collapse
Affiliation(s)
- Yuanchang Zhu
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Qing Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Meijian Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Tonghua Wu
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Weijie Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Ziqiang Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Bing Li
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Shikuan Zhang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Songmao Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Weidong Xie
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China
| | - Yuyang Jiang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Burton B Yang
- Sunnybrook Research Institute and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China.
| |
Collapse
|
67
|
Dixit A, Karande AA. Glycodelin regulates the numbers and function of peripheral natural killer cells. J Reprod Immunol 2019; 137:102625. [PMID: 31730930 DOI: 10.1016/j.jri.2019.102625] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/22/2019] [Accepted: 10/12/2019] [Indexed: 12/25/2022]
Abstract
Natural killer (NK) cells comprise of ∼70% of the immune cell population in the maternal decidua and ∼15% of the mononuclear cells in the peripheral blood. The decidual NK cells capable of producing high levels of cytokines are functionally distinct from the peripheral NK cells that exhibit high cytotoxicity. The numbers of peripheral NK cells and their cytotoxicity potential have been correlated with pregnancy outcome. In the same context, glycodelin, an immunomodulatory protein, has been recognized to be essential for the establishment and maintenance of pregnancy, and its' reduced levels are associated with recurrent spontaneous abortions. We investigated the effect of glycodelin on the peripheral NK cells. Our results reveal that glycodelin suppresses the cytotoxicity of peripheral NK cells via downregulating perforin, granzyme B and IFNγ. Glycodelin also induces caspase-dependent death in only activated peripheral NK cells, the effect suggested to be mediated by glycodelin upon engaging with the CD7 cell surface receptor. Thus, during pregnancy, glycodelin modulates the function and the number of cytotoxic NK cells that pose a deleterious effect on the fetus, a semi-allograft. This study provides insights into the mechanism of the regulatory effect of glycodelin on NK cells and could possibly be exploited for the management of miscarriages.
Collapse
Affiliation(s)
- Akanksha Dixit
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Anjali A Karande
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
68
|
Bortolotti D, Gentili V, Rotola A, Cultrera R, Marci R, Di Luca D, Rizzo R. HHV-6A infection of endometrial epithelial cells affects immune profile and trophoblast invasion. Am J Reprod Immunol 2019; 82:e13174. [PMID: 31338899 DOI: 10.1111/aji.13174] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022] Open
Abstract
PROBLEM We first reported human herpesvirus (HHV)-6A DNA presence in 43% of endometrial cells from women with idiopathic infertility, whereas no fertile control women harbored the virus. We investigated the effect of HHV-6A infection on the immunological status of the endometrium. METHOD OF STUDY Endometrial biopsies, uterine flushing, and whole blood samples were collected from 67 idiopathic infertile women (mid-secretory phase). We analyzed the endometrial immunological status evaluating: (a) the effect of HHV-6A infection on endometrial immune profile analyzing the ratio of interleukin (IL)-15/ fibroblast growth factor-inducible 14 (Fn-14) and IL-18/ TNF-related weak inducer of apoptosis (TWEAK) mRNA as a biomarker of endometrial (e)natural killer activation/maturation, angiogenesis, and Th1/Th2 balance; (b) endometrial receptivity to trophoblasts in endometrial 3D in vitro model; (c) natural killer (NK) cells and T cells percentage and subpopulations by flow cytometry. RESULTS We confirmed the presence of HHV-6A infection in a 40% of idiopathic infertile women, characterized by an immune profile reflecting eNK cell cytotoxic activation and a decrease in CD4+ CD25+ CD127dim/- regulatory T cells. The co-culture of endometrial epithelial cells with spheroids generated from the extravillous trophoblast-derived cell line JEG3 showed a twofold expansion of spheroids on endometrial epithelial-stromal cells (ESC) culture surface from HHV-6A negative women while no expansion was observed on the surface of ESC from HHV-6A positive women. CONCLUSION The identification of an effect of HHV-6A infection on endometrial immune status opens new perspectives in idiopathic infertile women care management. In addition, it would be possible to select antiviral therapies as novel, non-hormonal therapeutic approaches to those idiopathic infertile women characterized by the presence of endometrial HHV-6A infection, to increase their pregnancy rate.
Collapse
Affiliation(s)
- Daria Bortolotti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Valentina Gentili
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonella Rotola
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| | - Rosario Cultrera
- Department of Medical Sciences, Section of Dermathology and Infective Medicine, University of Ferrara, Ferrara, Italy
| | - Roberto Marci
- Department of Morphology, Surgery and Experimental Medicine, Section of Orthopedics, Obstetrics and Gynecology and Anesthesiology and Reanimation Ferrara, University of Ferrara, Ferrara, Italy.,School of Medicine, University of Geneva, Geneva, Switzerland
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberta Rizzo
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
69
|
Wu J, Gao FX, Wang C, Qin M, Han F, Xu T, Hu Z, Long Y, He XM, Deng X, Ren DL, Dai TY. IL-6 and IL-8 secreted by tumour cells impair the function of NK cells via the STAT3 pathway in oesophageal squamous cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:321. [PMID: 31324197 PMCID: PMC6642486 DOI: 10.1186/s13046-019-1310-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Background Recurrence and metastasis are the leading causes of tumour-related death in patients with oesophageal squamous cell carcinoma (ESCC). Tumour-infiltrating natural killer cells (NK cells) display powerful cytotoxicity to tumour cells and play a pivotal role in tumour therapy. However, the phenotype and functional regulation of NK cells in oesophageal squamous cell carcinoma (ESCC) remains largely unknown. Methods Single cell suspensions from blood and tissue samples were isolated by physical dissociation and filtering through a 70 μm cell strainer. Flow cytometry was applied to profile the activity and function of NK cells, and an antibody chip experiment was used to identify and quantitate cytokine levels. We studied IL-6 and IL-8 function in primary oesophageal squamous carcinoma and NK cell co-cultures in vitro and by a xenograft tumour model in vivo. Western blotting was used to quantitate STAT3 (signal transducer and activator of transcription 3) and p-STAT3 levels. Finally, we performed an IHC array to analyse IL-6/IL-8 (interleukin 6/interleukin 8) expression in 103 pairs of tumours and matched adjacent tissues of patients with ESCC to elucidate the correlation between IL-6 or IL-8 and clinical characteristics. Results The percentages of NK cells in both peripheral blood and tumour tissues from patients with ESCC were significantly increased in comparison with those in the controls and correlated with the clinical characteristics. Furthermore, the decrease in activating receptors and increase in inhibitory receptors on the surface of tumour-infiltrating NK cells was confirmed by flow cytometry. The level of granzyme B, the effector molecule of tumour-infiltrating NK cells, was also decreased. Mechanistically, primary ESCC cells activated the STAT3 signalling pathway on NK cells through IL-6 and IL-8 secretion, leading to the downregulation of activating receptors (NKp30 and NKG2D) on the surface of NK cells. An ex vivo study showed that blockade of STAT3 attenuated the IL-6/IL-8-mediated impairment of NK cell function. Moreover, the expression of IL-6 or IL-8 in tumour tissues was validated by immunohistochemistry to be positively correlated with tumour progression and poor survival, respectively. Conclusions Tumour cell-secreted IL-6 and IL-8 impair the activity and function of NK cells via STAT3 signalling and contribute to oesophageal squamous cell carcinoma malignancy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1310-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian Wu
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Feng-Xia Gao
- Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China
| | - Chao Wang
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Mei Qin
- Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China
| | - Fei Han
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Tao Xu
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Zhi Hu
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Yang Long
- Experimental Medicine Center, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Xue-Mei He
- Experimental Medicine Center, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China
| | - Xin Deng
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - De-Lian Ren
- Department of Immunology, Basic Medicine College, South West Medical University, Luzhou, Sichuan, China.
| | - Tian-Yang Dai
- Department of Thoracic Surgery, The Affiliated Hospital of South West Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
70
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
71
|
Brown JA, Singh G, Acklin JA, Lee S, Duehr JE, Chokola AN, Frere JJ, Hoffman KW, Foster GA, Krysztof D, Cadagan R, Jacobs AR, Stramer SL, Krammer F, García-Sastre A, Lim JK. Dengue Virus Immunity Increases Zika Virus-Induced Damage during Pregnancy. Immunity 2019; 50:751-762.e5. [PMID: 30737148 DOI: 10.1016/j.immuni.2019.01.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/30/2018] [Accepted: 01/10/2019] [Indexed: 01/13/2023]
Abstract
Zika virus (ZIKV) has recently been associated with birth defects and pregnancy loss after maternal infection. Because dengue virus (DENV) and ZIKV co-circulate, understanding the role of antibody-dependent enhancement in the context of pregnancy is critical. Here, we showed that the presence of DENV-specific antibodies in ZIKV-infected pregnant mice significantly increased placental damage, fetal growth restriction, and fetal resorption. This was associated with enhanced viral replication in the placenta that coincided with an increased frequency of infected trophoblasts. ZIKV-infected human placental tissues also showed increased replication in the presence of DENV antibodies, which was reversed by FcγR blocking antibodies. Furthermore, ZIKV-mediated fetal pathogenesis was enhanced in mice in the presence of a DENV-reactive monoclonal antibody, but not in the presence of the LALA variant, indicating a dependence on FcγR engagement. Our data suggest a possible mechanism for the recent increase in severe pregnancy outcomes after ZIKV infection in DENV-endemic areas.
Collapse
Affiliation(s)
- Julia A Brown
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gursewak Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua A Acklin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Silviana Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James E Duehr
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anupa N Chokola
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin J Frere
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin W Hoffman
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Richard Cadagan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam R Jacobs
- Department of Obstetrics and Gynecology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
72
|
|
73
|
Xu H, Du Y, He J, Wang L, Sun G. MicroRNA-378 protects human umbilical vein endothelial cells from injuries by soluble CD226 through down-regulating the expression of soluble CD226 in natural killer cells. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1640075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Huiying Xu
- Department of Obstetrics and Gynecology, The First People’s Hospital of Lanzhou City, Lanzhou, P.R. China
| | - Yu Du
- Department of Obstetrics and Gynecology, The First People’s Hospital of Lanzhou City, Lanzhou, P.R. China
| | - Jing He
- Department of Obstetrics and Gynecology, The First People’s Hospital of Lanzhou City, Lanzhou, P.R. China
| | - Liping Wang
- Department of Obstetrics and Gynecology, The First People’s Hospital of Lanzhou City, Lanzhou, P.R. China
| | - Gaogao Sun
- Department of Obstetrics and Gynecology, The First People’s Hospital of Lanzhou City, Lanzhou, P.R. China
| |
Collapse
|
74
|
Pollheimer J, Vondra S, Baltayeva J, Beristain AG, Knöfler M. Regulation of Placental Extravillous Trophoblasts by the Maternal Uterine Environment. Front Immunol 2018; 9:2597. [PMID: 30483261 PMCID: PMC6243063 DOI: 10.3389/fimmu.2018.02597] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
During placentation invasive extravillous trophoblasts (EVTs) migrate into the maternal uterus and modify its vessels. In particular, remodeling of the spiral arteries by EVTs is critical for adapting blood flow and nutrient transport to the developing fetus. Failures in this process have been noticed in different pregnancy complications such as preeclampsia, intrauterine growth restriction, stillbirth, or recurrent abortion. Upon invasion into the decidua, the endometrium of pregnancy, EVTs encounter different maternal cell types such as decidual macrophages, uterine NK (uNK) cells and stromal cells expressing a plethora of growth factors and cytokines. Here, we will summarize development of the EVT lineage, a process occurring independently of the uterine environment, and formation of its different subtypes. Further, we will discuss interactions of EVTs with arteries, veins and lymphatics and illustrate how the decidua and its different immune cells regulate EVT differentiation, invasion and survival. The present literature suggests that the decidual environment and its soluble factors critically modulate EVT function and reproductive success.
Collapse
Affiliation(s)
- Jürgen Pollheimer
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Vondra
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Jennet Baltayeva
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Guillermo Beristain
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
75
|
Li J, Tong C, Xu P, Wang L, Han TL, Wen L, Luo X, Tan B, Zhu F, Gui S, Gao R, Qi H, Baker PN. QSOX1 regulates trophoblastic apoptosis in preeclampsia through hydrogen peroxide production. J Matern Fetal Neonatal Med 2018; 32:3708-3715. [PMID: 29712536 DOI: 10.1080/14767058.2018.1471459] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Objective: Oxidative stress plays a significant role in the pathogenesis of preeclampsia (PE), by inducing trophoblast cell death and consequent placental dysfunction. Quiescin sulfhydryl oxidase 1 (QSOX1) is upregulated in many types of cancer cells; it promotes disulfide bond formation as well as hydrogen peroxide (H2O2) production. The aims of present study are to investigate the expression pattern of QSOX1 in placentae of pregnancies complicated by PE and the role of QSOX1 in the regulation of trophoblastic function, thus providing in-depth understanding of the putative involvement of QSOX1 in the development of PE. Methods: Human term placenta from normal pregnancies and from pregnancies complicated by PE was collected to measure QSOX1 expression and H2O2 levels. Down-regulation of QSOX1 in HTR-8/SVneo cells was achieved by siRNA interference. An in vitro cellular PE model was generated by hypoxic incubation. Protein expression levels were assessed by Western blotting, and H2O2 levels were determined in the cell culture medium as well as in the cell lysate. Trophoblast apoptosis was evaluated by TUNEL staining. Results: QSOX1 was overexpressed in the PE placenta. Inhibition of QSOX1 expression in HTR-8/SVneo cells attenuated cell apoptosis and intracellular H2O2 levels. Hypoxia-induced QSOX1 expression in HTR-8/SVneo cells and led to apoptosis of HTR-8/SVneo cells, and knock-down of QSOX1 rescued hypoxia-induced trophoblast apoptosis. Conclusions: Hypoxia-induced upregulation of QSOX1 and a consequent elevation in intracellular H2O2 increased apoptosis in placentae of pregnancies complicated by PE.
Collapse
Affiliation(s)
- Jinjin Li
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,c State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Chao Tong
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,c State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Ping Xu
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,c State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Lianlian Wang
- d Department of Reproduction Health and Infertility , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Ting-Li Han
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,e Liggins Institute, University of Auckland , Auckland , New Zealand
| | - Li Wen
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China
| | - Xiaofang Luo
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China
| | - Bin Tan
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China
| | - Fangyu Zhu
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China
| | - Shunping Gui
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China
| | - Rufei Gao
- b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,f Laboratory of Reproductive Biology, School of Public Health and Management , Chongqing Medical University , Chongqing , China
| | - Hongbo Qi
- a Department of Obstetrics , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China.,b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,c State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality , the First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Philip N Baker
- b International Collaborative Joint Laboratory of Reproduction and Development of Ministry of Education of China , Chongqing Medical University , Chongqing , China.,e Liggins Institute, University of Auckland , Auckland , New Zealand.,g College of Medicine, Biological Sciences and Psychology , University of Leicester , Leicester , United Kingdom
| |
Collapse
|
76
|
Rölle A, Meyer M, Calderazzo S, Jäger D, Momburg F. Distinct HLA-E Peptide Complexes Modify Antibody-Driven Effector Functions of Adaptive NK Cells. Cell Rep 2018; 24:1967-1976.e4. [PMID: 30134159 DOI: 10.1016/j.celrep.2018.07.069] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/25/2018] [Accepted: 07/18/2018] [Indexed: 11/29/2022] Open
Abstract
Adaptive NK cells are characterized by profound alterations in multiple signaling molecules, transcription factors, and epigenetic modifications compared with canonical NK cells. Although their existence is associated with prior exposure to human cytomegalovirus (HCMV), key questions regarding their regulation and function remain. A large proportion of adaptive NK cells express the activating receptor CD94/NKG2C, binding to human leukocyte antigen E (HLA-E), that presents a limited set of peptides. We show that adaptive NK cells discriminate differences between HLA-E-peptide complexes with exquisite specificity. Prolonged exposure to an environment displaying the HLA-E peptide ligand VMAPRTLFL, derived from the leader sequence of HLA-G, enriched adaptive NK cells with low FcεRγ expression, upregulated CD25 expression, increased proliferative activity, and resulted in elevated antibody-dependent cellular cytotoxicity and IFN-γ responses compared with other HLA-E peptide complexes. Our study demonstrates that recognition of alterations in the HLA-E ligandome via an activating receptor can influence heterologous effector mechanisms and proliferation in adaptive NK cells.
Collapse
Affiliation(s)
- Alexander Rölle
- Antigen Presentation and T/NK Cell Activation Group (D121), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor Immunity" (D120), German Cancer Research Center, Heidelberg, Germany.
| | - Marten Meyer
- Antigen Presentation and T/NK Cell Activation Group (D121), German Cancer Research Center (DKFZ), Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor Immunity" (D120), German Cancer Research Center, Heidelberg, Germany
| | - Silvia Calderazzo
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor Immunity" (D120), German Cancer Research Center, Heidelberg, Germany
| | - Frank Momburg
- Antigen Presentation and T/NK Cell Activation Group (D121), German Cancer Research Center (DKFZ), Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor Immunity" (D120), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
77
|
GPIbα is required for platelet-mediated hepatic thrombopoietin generation. Blood 2018; 132:622-634. [PMID: 29794068 DOI: 10.1182/blood-2017-12-820779] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Thrombopoietin (TPO), a hematopoietic growth factor produced predominantly by the liver, is essential for thrombopoiesis. Prevailing theory posits that circulating TPO levels are maintained through its clearance by platelets and megakaryocytes via surface c-Mpl receptor internalization. Interestingly, we found a two- to threefold decrease in circulating TPO in GPIbα-/- mice compared with wild-type (WT) controls, which was consistent in GPIbα-deficient human Bernard-Soulier syndrome (BSS) patients. We showed that lower TPO levels in GPIbα-deficient conditions were not due to increased TPO clearance by GPIbα-/- platelets but rather to decreased hepatic TPO mRNA transcription and production. We found that WT, but not GPIbα-/-, platelet transfusions rescued hepatic TPO mRNA and circulating TPO levels in GPIbα-/- mice. In vitro hepatocyte cocultures with platelets or GPIbα-coupled beads further confirm the disruption of platelet-mediated hepatic TPO generation in the absence of GPIbα. Treatment of GPIbα-/- platelets with neuraminidase caused significant desialylation; however, strikingly, desialylated GPIbα-/- platelets could not rescue impaired hepatic TPO production in vivo or in vitro, suggesting that GPIbα, independent of platelet desialylation, is a prerequisite for hepatic TPO generation. Additionally, impaired hepatic TPO production was recapitulated in interleukin-4/GPIbα-transgenic mice, as well as with antibodies targeting the extracellular portion of GPIbα, demonstrating that the N terminus of GPIbα is required for platelet-mediated hepatic TPO generation. These findings reveal a novel nonredundant regulatory role for platelets in hepatic TPO homeostasis, which improves our understanding of constitutive TPO regulation and has important implications in diseases related to GPIbα, such as BSS and auto- and alloimmune-mediated thrombocytopenias.
Collapse
|
78
|
Nurden AT. Acquired Antibodies to αIIbβ3 in Glanzmann Thrombasthenia: From Transfusion and Pregnancy to Bone Marrow Transplants and Beyond. Transfus Med Rev 2018; 32:S0887-7963(18)30037-3. [PMID: 29884513 DOI: 10.1016/j.tmrv.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/14/2018] [Accepted: 05/20/2018] [Indexed: 11/23/2022]
Abstract
Patients with the inherited bleeding disorder Glanzmann thrombasthenia (GT) possess platelets that lack αIIbβ3 integrin and fail to aggregate, and have moderate to severe mucocutaneous bleeding. Many become refractory to platelet transfusions due to the formation of isoantibodies to αIIbβ3 with the rapid elimination of donor platelets and/or a block of function. Epitope characterization has shown isoantibodies to be polyclonal and to recognize different epitopes on the integrin with β3 a major site and αvβ3 on endothelial and vascular cells a newly recognized target. Pregnancy in GT can also lead to isoantibody formation when fetal cells with β3 integrins pass into the circulation of a mother lacking them; a consequence is neonatal thrombocytopenia and a high risk of mortality. Antibody removal prior to donor transfusions can provide transient relief, but all evidence points to recombinant FVIIa as the first choice for GT patients either to stop bleeding or as prophylaxis. Promoting thrombin generation by rFVIIa favors GT platelet interaction with fibrin, and the risk of deep vein thrombosis also associated with prolonged immobilization and catheter use requires surveillance. Although having a high risk, allogeneic bone marrow transplantation associated with different stem cell sources and conditioning regimens has proved successful in many cases of severe GT with antibodies, and often, the associated conditioning and immunosuppressive therapy leads to loss of isoantibody production. Animal models of gene therapy for GT show promising results, but isoantibody production can be stimulated and CRISPR/Cas9 technology has yet to be applied. Up-to-date consensus protocols for dealing with isoantibodies in GT are urgently required, and networks providing patient care should be expanded.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
79
|
Castellana B, Perdu S, Kim Y, Chan K, Atif J, Marziali M, Beristain AG. Maternal obesity alters uterine NK activity through a functional KIR2DL1/S1 imbalance. Immunol Cell Biol 2018; 96:805-819. [PMID: 29569748 DOI: 10.1111/imcb.12041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/28/2022]
Abstract
In pregnancy, uterine natural killer cells (uNK) play essential roles in coordinating uterine angiogenesis, blood vessel remodeling and promoting maternal tolerance to fetal tissue. Deviances from a normal uterine microenvironment are thought to modify uNK function(s) by limiting their ability to establish a healthy pregnancy. While maternal obesity has become a major health concern due to associations with adverse effects on fetal and maternal health, our understanding into how obesity contributes to poor pregnancy disorders is unknown. Given the importance of uNK in pregnancy, this study examines the impact of obesity on uNK function in women in early pregnancy. We identify that uNK from obese women show a greater propensity for cellular activation, but this difference does not translate into increased effector killing potential. Instead, uNK from obese women express an altered repertoire of natural killer receptors, including an imbalance in inhibitory KIR2DL1 and activating KIR2DS1 receptors that favors HLA-C2-directed uNK activation. Notably, we show that obesity-related KIR2DS1 skewing potentiates TNFα production upon receptor crosslinking. Together, these findings suggest that maternal obesity modifies uNK activity by altering the response toward HLA-C2 antigen and KIR2DL1/2DS1-controlled TNFα release. Furthermore, this work identifies alterations in uNK function resulting from maternal obesity that may impact early developmental processes important in pregnancy health.
Collapse
Affiliation(s)
- Barbara Castellana
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Sofie Perdu
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Yoona Kim
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kathy Chan
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Jawairia Atif
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Megan Marziali
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Alexander G Beristain
- British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|