1
|
Soucek O, Kacerovsky M, Kacerovska Musilova I, Stranik J, Kukla R, Bolehovska R, Andrys C. Absolute counts of leukocyte subsets in peripheral blood in pregnancies complicated by preterm prelabour rupture of membranes. J OBSTET GYNAECOL 2024; 44:2390575. [PMID: 39157927 DOI: 10.1080/01443615.2024.2390575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND This study aimed to assess variations in the absolute counts of various leukocyte subsets in the peripheral blood of women with pregnancies affected by preterm prelabour rupture of membranes (PPROM), in relation to the presence of intra-amniotic inflammation (IAI). METHODS The study included fifty-two women with singleton pregnancies experiencing PPROM. Absolute counts of different leukocyte subpopulations, such as granulocytes, monocytes, lymphocytes, T cells and their subsets, B cells and their subsets, and NK cells and their subsets, were measured in maternal peripheral blood samples using multicolour flow cytometry. IAI was identified by elevated concentrations of interleukin 6 (IL-6) in the amniotic fluid, which was collected through transabdominal amniocentesis. RESULTS Women with IAI exhibited higher absolute counts of leukocytes (p = 0.003), granulocytes (p = 0.008), and monocytes (p = 0.009). However, the presence of IAI did not significantly affect the absolute counts of lymphocytes or their subpopulations. CONCLUSIONS The study found that IAI is associated with changes in the absolute counts of leukocytes from the innate immunity compartment in the peripheral blood of women with pregnancies complicated by PPROM. Conversely, it does not significantly alter the counts of cells from the adaptive immune system. The changes observed may reflect the natural, temporal, and localised characteristics of IAI.
Collapse
Affiliation(s)
- Ondrej Soucek
- Department of Clinical Immunology and Allergy, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, Hospital Most, Most, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ivana Kacerovska Musilova
- Department of Obstetrics and Gynecology, Hospital Most, Most, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Rudolf Kukla
- Department of Microbiology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Radka Bolehovska
- Department of Microbiology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ctirad Andrys
- Department of Clinical Immunology and Allergy, Charles University in Prague, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
2
|
Padron JG, Saito Reis CA, Ng PK, Norman Ing ND, Baker H, Davis K, Kurashima C, Kendal-Wright CE. Stretch Causes cffDNA and HMGB1-Mediated Inflammation and Cellular Stress in Human Fetal Membranes. Int J Mol Sci 2024; 25:5161. [PMID: 38791199 PMCID: PMC11121497 DOI: 10.3390/ijms25105161] [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: 03/26/2024] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Danger-associated molecular patterns (DAMPs) are elevated within the amniotic cavity, and their increases correlate with advancing gestational age, chorioamnionitis, and labor. Although the specific triggers for their release in utero remain unclear, it is thought that they may contribute to the initiation of parturition by influencing cellular stress mechanisms that make the fetal membranes (FMs) more susceptible to rupture. DAMPs induce inflammation in many different tissue types. Indeed, they precipitate the subsequent release of several proinflammatory cytokines that are known to be key for the weakening of FMs. Previously, we have shown that in vitro stretch of human amnion epithelial cells (hAECs) induces a cellular stress response that increases high-mobility group box-1 (HMGB1) secretion. We have also shown that cell-free fetal DNA (cffDNA) induces a cytokine response in FM explants that is fetal sex-specific. Therefore, the aim of this work was to further investigate the link between stretch and the DAMPs HMGB1 and cffDNA in the FM. These data show that stretch increases the level of cffDNA released from hAECs. It also confirms the importance of the sex of the fetus by demonstrating that female cffDNA induced more cellular stress than male fetuses. Our data treating hAECs and human amnion mesenchymal cells with HMGB1 show that it has a differential effect on the ability of the cells of the amnion to upregulate the proinflammatory cytokines and propagate a proinflammatory signal through the FM that may weaken it. Finally, our data show that sulforaphane (SFN), a potent activator of Nrf2, is able to mitigate the proinflammatory effects of stretch by decreasing the levels of HMGB1 release and ROS generation after stretch and modulating the increase of key cytokines after cell stress. HMGB1 and cffDNA are two of the few DAMPs that are known to induce cytokine release and matrix metalloproteinase (MMP) activation in the FMs; thus, these data support the general thesis that they can function as potential central players in the normal mechanisms of FM weakening during the normal distension of this tissue at the end of a normal pregnancy.
Collapse
Affiliation(s)
- Justin Gary Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96822, USA;
- Wayne State School of Medicine, Detroit, MI 48201, USA
| | - Chelsea A. Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Po’okela K. Ng
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Nainoa D. Norman Ing
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Hannah Baker
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Kamalei Davis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Courtney Kurashima
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Claire E. Kendal-Wright
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96822, USA;
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
- Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96826, USA
| |
Collapse
|
3
|
Flores-Espinosa P, Mancilla-Herrera I, Olmos-Ortiz A, Díaz L, Zaga-Clavellina V. Evaluation of Leukocyte Chemotaxis Induced by Human Fetal Membranes in an In Vitro Model. Methods Mol Biol 2024; 2781:27-37. [PMID: 38502440 DOI: 10.1007/978-1-0716-3746-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Leukocyte infiltration into the maternal-fetal interface is a consequence of the robust inflammation in the gestational tissues during term labor and preterm labor with or without infection. During pregnancy, the fetal membranes act as a physical barrier that isolates the fetus into the amniotic cavity, keeping it in an optimal environment for its development. In addition, the fetal membranes possess immunological competencies such as the secretion of cytokines and chemokines in response to different stimuli. Clinical and experimental evidence indicates that these tissues are involved in the extensive chemotaxis of immune cells in normal or pathological conditions.Few studies have evaluated the chemotactic capacities of the fetal membranes considering that this tissue is composed of two adjacent tissues, the amnion and the chorion, which have different characteristics. Although these tissues function as a unit, their response is complex since there is an interaction between them, where each tissue contributes differently. The protocol described here allows us to evaluate the in vitro chemotactic capacities of fetal membranes in response to various applied stimuli, considering the contribution of each of their components (amnion and choriodecidua) using a Boyden chamber assay and phenotyping the chemo-attracted leukocytes by flow cytometry.
Collapse
Affiliation(s)
- Pilar Flores-Espinosa
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico.
| | - Ismael Mancilla-Herrera
- Department of Infectology and Immunology, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Andrea Olmos-Ortiz
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, , Mexico City, Mexico
| | - Verónica Zaga-Clavellina
- Department of Immunobiochemistry, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| |
Collapse
|
4
|
Shynlova O, Nadeem L, Lye S. Progesterone control of myometrial contractility. J Steroid Biochem Mol Biol 2023; 234:106397. [PMID: 37683774 DOI: 10.1016/j.jsbmb.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
During pregnancy, the primary function of the uterus is to be quiescent and not contract, which allows the growing fetus to develop and mature. A uterine muscle layer, myometrium, is composed of smooth muscle cells (SMCs). Before the onset of labor contractions, the uterine SMCs experience a complex biochemical and molecular transformation involving the expression of contraction-associated proteins. Labor is initiated when genes in SMCs are activated in response to a combination of hormonal, inflammatory and mechanical signals. In this review, we provide an overview of molecular mechanisms regulating the process of parturition in humans, focusing on the hormonal control of the myometrium, particularly the steroid hormone progesterone. The primary reason for discussing the regulation of myometrial contractility by progesterone is the importance of the clinical problem of preterm birth. It is thought that the hormonal mechanisms regulating premature uterine contractions represent an untimely triggering of the normal events occurring during term parturition. Yet, our knowledge of the complex and redundant hormonal pathways controlling uterine contractile activity leading to delivery of the neonate remains incomplete. Finally, we introduce recent animal studies using a novel class of drugs, Selective Progesterone Receptor Modulators, targeting progesterone signaling to prevent premature myometrial contractions.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada
| |
Collapse
|
5
|
Wanvimonsuk S, Somboonwiwat K. Peroxiredoxin-4 supplementation modulates the immune response, shapes the intestinal microbiome, and enhances AHPND resistance in Penaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2023:108915. [PMID: 37355217 DOI: 10.1016/j.fsi.2023.108915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
Peroxiredoxin-4 from Penaeus vannamei (LvPrx4) is considered a damage-associated molecular pattern (DAMP) that can activate the expression of immune-related genes through the Toll pathway. We previously demonstrated that the recombinant LvPrx4 (rLvPrx4) can enhance shrimp resistance against Vibrio parahaemolyticus, causing acute hepatopancreatic necrosis disease (VPAHPND), which causes great production losses in shrimp farming. Herein, we showed that the rLvPrx4 had a thermal tolerance of around 60 °C and that the ionic strength had no noticeable effect on its activity. We discovered that feeding a diet containing rLvPrx4 to shrimp for three weeks increased the expression of the immune-related genes LvPEN4 and LvVago5. Furthermore, pre-treatment with rLvPrx4 feeding could significantly prolong shrimp survival following the VPAHPND challenge. The shrimp intestinal microbiome was then characterized using PCR amplification of the 16S rRNA gene and Illumina sequencing. Three weeks of rLvPrx4 supplementation altered the bacterial community structure (beta diversity) and revealed the induction of differentially abundant families, including Cryomorphaceae, Flavobacteriaceae, Pirellulaceae, Rhodobacteraceae, and Verrucomicrobiaceae, in the rLvPrx4 group. Metagenomic predictions indicated that some amino acid metabolism pathways, such as arginine and proline metabolism, and genetic information processing were significantly elevated in the rLvPrx4 group compared to the control group. This study is the first to describe the potential use of rLvPrx4 supplementation to enhance shrimp resistance to VPAHPND and alter the composition of a beneficial bacterial community in shrimp, making rLvPrx4 a promising feed supplement as an alternative to antibiotics for controlling VPAHPND infection in shrimp aquaculture.
Collapse
Affiliation(s)
- Supitcha Wanvimonsuk
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kunlaya Somboonwiwat
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
6
|
Guo X, Wang Y, Yu H. Relationship between placental pathology and neonatal outcomes. Front Pediatr 2023; 11:1201991. [PMID: 37397153 PMCID: PMC10309182 DOI: 10.3389/fped.2023.1201991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Objective To evaluate the relationship between maternal vascular malperfusion and acute intrauterine infection/inflammation with neonatal outcomes. Methods This was a retrospective study of women with singleton pregnancies who completed placenta pathological examination. The aim was to study the distribution of acute intrauterine infection/inflammation and maternal placental vascular malperfusion among groups with preterm birth and/or rupture of membranes. The relationship between two subtypes of placental pathology and neonatal gestational age, birth weight Z-score, neonatal respiratory distress syndrome, and intraventricular hemorrhage was further explored. Results 990 pregnant women were divided into four groups, including 651 term, 339 preterm, 113 women with premature rupture of membranes, and 79 with preterm premature rupture of membranes. The incidence of respiratory distress syndrome and intraventricular hemorrhage in four groups were (0.7%, 0.0%, 31.9%, 31.6%, P < 0.001) and (0.9%, 0.9%, 20.0%, 17.7%, P < 0.001), respectively. The incidence of maternal vascular malperfusion and acute intrauterine infection/inflammation were (82.0%, 77.0%, 75.8%, 72.1%, P = 0.06) and (21.9%, 26.5%, 23.1%, 44.3%, P = 0.010), respectively. Acute intrauterine infection/inflammation was associated with shorter gestational age (adjusted difference -4.7 weeks, P < 0.001) and decreased weight (adjusted Z score -2.6, P < 0.001) than those with no lesions in preterm birth. When two subtype placenta lesions co-occurrence, shorter gestational age (adjusted difference -3.0 weeks, P < 0.001) and decreased weight (adjusted Z score -1.8, P < 0.001) were observed in preterm. Consistent findings were observed in preterm births with or without premature rupture of membranes. In addition, acute infection/inflammation and maternal placenta malperfusion alone or in combination were associated with an increased risk of neonatal respiratory distress syndrome (adjusted odds ratio (aOR) 0.8, 1.5, 1.8), but the difference was not statistically significant. Conclusion Maternal vascular malperfusion and acute intrauterine infection/inflammation alone or co-occurrence are associated with adverse neonatal outcomes, which may provide new ideas for clinical diagnosis and treatment.
Collapse
|
7
|
Flores-Espinosa P, Méndez I, Irles C, Olmos-Ortiz A, Helguera-Repetto C, Mancilla-Herrera I, Ortuño-Sahagún D, Goffin V, Zaga-Clavellina V. Immunomodulatory role of decidual prolactin on the human fetal membranes and placenta. Front Immunol 2023; 14:1212736. [PMID: 37359537 PMCID: PMC10288977 DOI: 10.3389/fimmu.2023.1212736] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
The close interaction between fetal and maternal cells during pregnancy requires multiple immune-endocrine mechanisms to provide the fetus with a tolerogenic environment and protection against any infectious challenge. The fetal membranes and placenta create a hyperprolactinemic milieu in which prolactin (PRL) synthesized by the maternal decidua is transported through the amnion-chorion and accumulated into the amniotic cavity, where the fetus is bedded in high concentrations during pregnancy. PRL is a pleiotropic immune-neuroendocrine hormone with multiple immunomodulatory functions mainly related to reproduction. However, the biological role of PRL at the maternal-fetal interface has yet to be fully elucidated. In this review, we have summarized the current information on the multiple effects of PRL, focusing on its immunological effects and biological significance for the immune privilege of the maternal-fetal interface.
Collapse
Affiliation(s)
- Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Isabel Méndez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Claudine Irles
- Institut National de la Santé et de la Recherche Médicale (INSERM) U978, Université Sorbonne Paris Nord, Unité de Formation et de Recherche (UFR) Santé Médecine et Biologie Humaine (SMBH), Bobigny, France
| | - Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Cecilia Helguera-Repetto
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Ismael Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara, Mexico
| | - Vincent Goffin
- Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche (UMR)-S1151, CNRS Unité Mixte de Recherche (UMR)-S8253, Institut Necker Enfants Malades, Paris, France
| | - Verónica Zaga-Clavellina
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México, Mexico
| |
Collapse
|
8
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther 2023; 8:207. [PMID: 37211559 DOI: 10.1038/s41392-023-01452-1] [Citation(s) in RCA: 312] [Impact Index Per Article: 312.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/06/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
Macrophages exist in various tissues, several body cavities, and around mucosal surfaces and are a vital part of the innate immune system for host defense against many pathogens and cancers. Macrophages possess binary M1/M2 macrophage polarization settings, which perform a central role in an array of immune tasks via intrinsic signal cascades and, therefore, must be precisely regulated. Many crucial questions about macrophage signaling and immune modulation are yet to be uncovered. In addition, the clinical importance of tumor-associated macrophages is becoming more widely recognized as significant progress has been made in understanding their biology. Moreover, they are an integral part of the tumor microenvironment, playing a part in the regulation of a wide variety of processes including angiogenesis, extracellular matrix transformation, cancer cell proliferation, metastasis, immunosuppression, and resistance to chemotherapeutic and checkpoint blockade immunotherapies. Herein, we discuss immune regulation in macrophage polarization and signaling, mechanical stresses and modulation, metabolic signaling pathways, mitochondrial and transcriptional, and epigenetic regulation. Furthermore, we have broadly extended the understanding of macrophages in extracellular traps and the essential roles of autophagy and aging in regulating macrophage functions. Moreover, we discussed recent advances in macrophages-mediated immune regulation of autoimmune diseases and tumorigenesis. Lastly, we discussed targeted macrophage therapy to portray prospective targets for therapeutic strategies in health and diseases.
Collapse
Affiliation(s)
- Shanze Chen
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Abdullah F U H Saeed
- Department of Cancer Biology, Beckman Research Institute of City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen University, Shenzhen, 518052, China
| | - Qiong Jiang
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Haizhao Xu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
- Department of Respiratory, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gary Guishan Xiao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Yanhong Duo
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Coste K, Bruet S, Chollat-Namy C, Filhol O, Cochet C, Gallot D, Marceau G, Blanchon L, Sapin V, Belville C. Characterization of RAGE and CK2 Expressions in Human Fetal Membranes. Int J Mol Sci 2023; 24:ijms24044074. [PMID: 36835482 PMCID: PMC9966553 DOI: 10.3390/ijms24044074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
At the feto-maternal interface, fetal membranes (FM) play a crucial role throughout pregnancy. FM rupture at term implicates different sterile inflammation mechanisms including pathways activated by the transmembrane glycoprotein receptor for advanced glycation end-products (RAGE) belonging to the immunoglobulin superfamily. As the protein kinase CK2 is also implicated in the inflammation process, we aimed to characterize the expressions of RAGE and the protein kinase CK2 as a candidate regulator of RAGE expression. The amnion and choriodecidua were collected from FM explants and/or primary amniotic epithelial cells throughout pregnancy and at term in spontaneous labor (TIL) or term without labor (TNL). The mRNA and protein expressions of RAGE and the CK2α, CK2α', and CK2β subunits were investigated using reverse transcription quantitative polymerase chain reaction and Western blot assays. Their cellular localizations were determined with microscopic analyses, and the CK2 activity level was measured. RAGE and the CK2α, CK2α', and CK2β subunits were expressed in both FM layers throughout pregnancy. At term, RAGE was overexpressed in the amnion from the TNL samples, whereas the CK2 subunits were expressed at the same level in the different groups (amnion/choriodecidua/amniocytes, TIL/TNL), without modification of the CK2 activity level and immunolocalization. This work paves the way for future experiments regarding the regulation of RAGE expression by CK2 phosphorylation.
Collapse
Affiliation(s)
- Karen Coste
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Neonatal Intensive Care Department, F-63000 Clermont-Ferrand, France
| | - Shaam Bruet
- CHU Clermont-Ferrand, Neonatal Intensive Care Department, F-63000 Clermont-Ferrand, France
| | - Caroline Chollat-Namy
- CHU Clermont-Ferrand, Neonatal Intensive Care Department, F-63000 Clermont-Ferrand, France
| | - Odile Filhol
- INSERM, CEA, UMR Biosanté, U1292, University Grenoble Alpes, F-38000 Grenoble, France
| | - Claude Cochet
- INSERM, CEA, UMR Biosanté, U1292, University Grenoble Alpes, F-38000 Grenoble, France
| | - Denis Gallot
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Obstetrics and Gynecology Department, F-63000 Clermont-Ferrand, France
| | - Geoffroy Marceau
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Biochemistry and Molecular Genetic Department, F-63000 Clermont-Ferrand, France
| | - Loïc Blanchon
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Vincent Sapin
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- CHU Clermont-Ferrand, Biochemistry and Molecular Genetic Department, F-63000 Clermont-Ferrand, France
| | - Corinne Belville
- iGReD, Team “Translational Approach to Epithelial Injury and Repair”, UMR6293 CNRS-U1103 INSERM, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
- Correspondence: ; Tel.: +33-4-7317-8174
| |
Collapse
|
10
|
Sudiarta KE, Candra CJ, Khan J, Rahadianto, Handajani F. RAGE and HMGB1 expressions in fetal membranes of premature rupture of membranes patients. MEDICAL JOURNAL OF INDONESIA 2022. [DOI: 10.13181/mji.oa.226099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Premature rupture of membranes (PROM) often occurs in pregnancy. The fetal membrane weakening is caused by inflammation involving receptor activation for advanced glycation end-products (RAGE) and high mobility group box protein 1 (HMGB1). The associations between RAGE and HMGB1 with PROM are rarely studied. Hence, this study aimed to determine those associations in fetal membranes with PROM occurrence.
METHODS This case-control study was conducted at Dr. Ramelan Central Naval Hospital, Surabaya, Indonesia, from August to November 2019. The subjects, determined using a non-probability sampling method (a saturated sample), were divided into PROM and normal pregnancy with intact fetal membranes (control) groups. Fetal membrane specimens were collected during vaginal and cesarean section deliveries. The expressions of RAGE and HMGB1 were determined using the immunohistochemical method and further analyzed using the Mann–Whitney U test.
RESULTS The expression of RAGE in fetal membranes with PROM was significantly higher than the control (52.74% versus 14.9% expression/mm2, p<0.001), as well as the expression of HMGB1 (45.9% versus 8.5% expression/mm2, p<0.001).
CONCLUSIONS The higher expressions of RAGE and HMGB1 in fetal membranes were associated with PROM.
Collapse
|
11
|
Cincotta AH, Cersosimo E, Alatrach M, Ezrokhi M, Agyin C, Adams J, Chilton R, Triplitt C, Chamarthi B, Cominos N, DeFronzo RA. Bromocriptine-QR Therapy Reduces Sympathetic Tone and Ameliorates a Pro-Oxidative/Pro-Inflammatory Phenotype in Peripheral Blood Mononuclear Cells and Plasma of Type 2 Diabetes Subjects. Int J Mol Sci 2022; 23:ijms23168851. [PMID: 36012132 PMCID: PMC9407769 DOI: 10.3390/ijms23168851] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Bromocriptine-QR is a sympatholytic dopamine D2 agonist for the treatment of type 2 diabetes that has demonstrated rapid (within 1 year) substantial reductions in adverse cardiovascular events in this population by as yet incompletely delineated mechanisms. However, a chronic state of elevated sympathetic nervous system activity and central hypodopaminergic function has been demonstrated to potentiate an immune system pro-oxidative/pro-inflammatory condition and this immune phenotype is known to contribute significantly to the advancement of cardiovascular disease (CVD). Therefore, the possibility exists that bromocriptine-QR therapy may reduce adverse cardiovascular events in type 2 diabetes subjects via attenuation of this underlying chronic pro-oxidative/pro-inflammatory state. The present study was undertaken to assess the impact of bromocriptine-QR on a wide range of immune pro-oxidative/pro-inflammatory biochemical pathways and genes known to be operative in the genesis and progression of CVD. Inflammatory peripheral blood mononuclear cell biology is both a significant contributor to cardiovascular disease and also a marker of the body’s systemic pro-inflammatory status. Therefore, this study investigated the effects of 4-month circadian-timed (within 2 h of waking in the morning) bromocriptine-QR therapy (3.2 mg/day) in type 2 diabetes subjects whose glycemia was not optimally controlled on the glucagon-like peptide 1 receptor agonist on (i) gene expression status (via qPCR) of a wide array of mononuclear cell pro-oxidative/pro-inflammatory genes known to participate in the genesis and progression of CVD (OXR1, NRF2, NQO1, SOD1, SOD2, CAT, GSR, GPX1, GPX4, GCH1, HMOX1, BiP, EIF2α, ATF4, PERK, XBP1, ATF6, CHOP, GSK3β, NFkB, TXNIP, PIN1, BECN1, TLR2, TLR4, TLR10, MAPK8, NLRP3, CCR2, GCR, L-selectin, VCAM1, ICAM1) and (ii) humoral measures of sympathetic tone (norepinephrine and normetanephrine), whole-body oxidative stress (nitrotyrosine, TBARS), and pro-inflammatory factors (IL-1β, IL-6, IL-18, MCP-1, prolactin, C-reactive protein [CRP]). Relative to pre-treatment status, 4 months of bromocriptine-QR therapy resulted in significant reductions of mRNA levels in PBMC endoplasmic reticulum stress-unfolded protein response effectors [GRP78/BiP (34%), EIF2α (32%), ATF4 (29%), XBP1 (25%), PIN1 (14%), BECN1 (23%)], oxidative stress response proteins [OXR1 (31%), NRF2 (32%), NQO1 (39%), SOD1 (52%), CAT (26%), GPX1 (33%), GPX4 (31%), GCH1 (30%), HMOX1 (40%)], mRNA levels of TLR pro-inflammatory pathway proteins [TLR2 (46%), TLR4 (20%), GSK3β (19%), NFkB (33%), TXNIP (18%), NLRP3 (32%), CCR2 (24%), GCR (28%)], mRNA levels of pro-inflammatory cellular receptor proteins CCR2 and GCR by 24% and 28%, and adhesion molecule proteins L-selectin (35%) and VCAM1 (24%). Relative to baseline, bromocriptine-QR therapy also significantly reduced plasma levels of norepinephrine and normetanephrine by 33% and 22%, respectively, plasma pro-oxidative markers nitrotyrosine and TBARS by 13% and 10%, respectively, and pro-inflammatory factors IL-18, MCP1, IL-1β, prolactin, and CRP by 21%,13%, 12%, 42%, and 45%, respectively. These findings suggest a unique role for circadian-timed bromocriptine-QR sympatholytic dopamine agonist therapy in reducing systemic low-grade sterile inflammation to thereby reduce cardiovascular disease risk.
Collapse
Affiliation(s)
- Anthony H. Cincotta
- VeroScience LLC, Tiverton, RI 02878, USA
- Correspondence: ; Tel.: +1-401-816-0525
| | - Eugenio Cersosimo
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Mariam Alatrach
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | - Christina Agyin
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - John Adams
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Chilton
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Curtis Triplitt
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | | | - Ralph A. DeFronzo
- Texas Diabetes Institute, University Health System, San Antonio, TX 78207, USA
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
12
|
Kroupa M, Tomasova K, Kavec M, Skrobanek P, Buchler T, Kumar R, Vodickova L, Vodicka P. TElomeric repeat-containing RNA (TERRA): Physiological functions and relevance in cancer. Front Oncol 2022; 12:913314. [PMID: 35982970 PMCID: PMC9380590 DOI: 10.3389/fonc.2022.913314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Telomeres are complex protective structures located at the ends of linear eukaryotic chromosomes. Their purpose is to prevent genomic instability. Research progress in telomere biology during the past decades has identified a network of telomeric transcripts of which the best-studied is TElomeric Repeat-containing RNA (TERRA). TERRA was shown to be important not only for the preservation of telomere homeostasis and genomic stability but also for the expression of hundreds of genes across the human genome. These findings added a new level of complexity to telomere biology. Herein we provide insights on the telomere transcriptome, its relevance for proper telomere function, and its implications in human pathology. We also discuss possible clinical opportunities of exosomal telomere transcripts detection as a biomarker in cancer precision medicine.
Collapse
Affiliation(s)
- Michal Kroupa
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czechia
- *Correspondence: Michal Kroupa, ; Pavel Vodicka,
| | - Kristyna Tomasova
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czechia
| | - Miriam Kavec
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Pavel Skrobanek
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, Prague, Czechia
| | - Rajiv Kumar
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Ludmila Vodickova
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czechia
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Pavel Vodicka
- Department of the Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czechia
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czechia
- *Correspondence: Michal Kroupa, ; Pavel Vodicka,
| |
Collapse
|
13
|
Xie X, Bai G, Zhang L, Liu H, Qiang D, Li L. Changes in plasma IRAK-M in patients with prediabetes and its relationship with related metabolic indexes: a cross-sectional study. J Int Med Res 2022; 50:3000605221111275. [PMID: 36039603 PMCID: PMC9437484 DOI: 10.1177/03000605221111275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate whether IL-1R-associated kinase (IRAK)-M is associated with prediabetes and type 2 diabetes (T2D). METHODS In this cross-sectional study, enrolled subjects were assigned to different groups according to their fasting plasma glucose (FPG) values. IRAK-M and metabolic parameters, including fasting insulin (FINS), glycosylated hemoglobin (HbA1c), homeostasis model assessment of insulin resistance (HOMA-IR) and beta-cell function (HOMA-β), and thioredoxin-interacting protein (TXNIP), were evaluated. The area under the receiver operating characteristic curve of IRAK-M and TXNIP for prediabetes and T2D was determined. RESULTS IRAK-M decreased significantly with increasing FPG levels. IRAK-M was negatively correlated with TXNIP, FPG, FINS, HbA1c, and HOMA-IR and positively correlated with HOMA-β. The diagnostic cutoff value of IRAK-M was 3.76 ng/mL for prediabetes and 3.45 ng/mL for T2D. After stratifying by IRAK-M (<3.76 and ≥3.76 ng/mL), patients with a higher TXNIP level showed a greater risk of prediabetes or T2D in the subgroup with low IRAK-M (<3.76 ng/mL). CONCLUSIONS IRAK-M is independently and positively associated with prediabetes and T2D, while TXNIP is independently and negatively associated with prediabetes and T2D. IRAK-M and TXNIP serve as diagnostic factors for prediabetes.
Collapse
Affiliation(s)
- Xiaomin Xie
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Guirong Bai
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Li Zhang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Huili Liu
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Dan Qiang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Ling Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, Ningxia, China
| |
Collapse
|
14
|
Richardson L, Menon R. Fetal membrane at the feto-maternal interface: An underappreciated and understudied intrauterine tissue. PLACENTA AND REPRODUCTIVE MEDICINE 2022; 1:10.54844/prm.2022.0104. [PMID: 37502422 PMCID: PMC10373051 DOI: 10.54844/prm.2022.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Affiliation(s)
- Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
| |
Collapse
|
15
|
Wan S, Chen P, Gu M, Liu J, Zhou Q, Zhang F, Lu Y, Li L, Wang X. Fetal Lung-Derived Exosomes in Term Labor Amniotic Fluid Induce Amniotic Membrane Senescence. Front Cell Dev Biol 2022; 10:889861. [PMID: 35859898 PMCID: PMC9289145 DOI: 10.3389/fcell.2022.889861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
The mechanism of parturition is still unclear. Evidence has shown that delivery is associated with cellular senescence of the amniotic membrane. We isolated fetal lung-associated exosomes from the amniotic fluid from term labor (TL-exos) and verified that the exosomes can cause primary human amniotic epithelial cell (hAEC) senescence and apoptosis and can release higher levels of senescence-associated secretory phenotype (SASP)-related molecules and proinflammatory damage-associated molecular patterns (DAMPs) than exosomes isolated from the amniotic fluid from term not in labor (TNIL-exos). The human lung carcinoma cell lines (A549) can be used as an alternative to alveolar type 2 epithelial cells producing pulmonary surfactant. Therefore, we isolated A549 cell-derived exosomes (A549-exos) and found that they can trigger hAEC to undergo the same aging process. Finally, the animal experiments suggested that A549-exos induced vaginal bleeding and preterm labor in pregnant mice. Therefore, we conclude that exosomes derived from fetal lungs in term labor amniotic fluid induce amniotic membrane senescence, which may provide new insight into the mechanism of delivery.
Collapse
Affiliation(s)
- Shuting Wan
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
| | - Mengqi Gu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
| | - Jing Liu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
| | - Qian Zhou
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
| | - Fengyuan Zhang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, China
| | - Yuan Lu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
- *Correspondence: Lei Li, ; Yuan Lu, ; Xietong Wang,
| | - Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, China
- *Correspondence: Lei Li, ; Yuan Lu, ; Xietong Wang,
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong University, Jinan, China
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, China
- Department of Obstetrics and Gynaecology, Maternal and Child Health Care of Shandong Province, Jinan, China
- The Laboratory of Placenta-Related Diseases, Key Laboratory of Birth Regulation and Control Technology of the National Health and Family Planning Commission of China, Jinan, China
- *Correspondence: Lei Li, ; Yuan Lu, ; Xietong Wang,
| |
Collapse
|
16
|
Saito Reis CA, Ng PK, Kurashima CK, Padron J, Kendal-Wright CE. Fetal DNA Causes Sex-Specific Inflammation From Human Fetal Membranes. Front Physiol 2022; 13:901726. [PMID: 35812324 PMCID: PMC9257279 DOI: 10.3389/fphys.2022.901726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Inflammation is central to the mechanisms of parturition, but the lack of understanding of how it is controlled in normal parturition hampers our ability to understand how it may diverge resulting in preterm birth. Cell-free fetal DNA is found in the amniotic fluid, and it is thought to be able to activate inflammation as a danger-associated molecular pattern. Although its levels increases with gestational age, its effect has not been studied on the human fetal membranes. Thus, the aim of this study was to determine if the fetal DNA can trigger inflammation in the human fetal membranes and, thus, potentially contribute to the inflammatory load. Isolated human amniotic epithelial cells and fetal membrane explants were treated apically with fetal DNA causing the translocation of NF-KB into the nucleus of cells and throughout the cells of the explant layers with time. Fetal membrane explants were treated apically with either small or larger fragments of fetal DNA. IL-6, TNFα, and GM-CSF secretion was measured by ELISA, and pro-MMP2 and pro-MMP9 activity was measured by zymography from apical and basal media. Increased apical IL-6 secretion and basal pro-MMP2 activity was seen with small fragments of fetal DNA. When the data were disaggregated based on fetal sex, males had significant increases in IL-6 secretion and basal increased activity in pro-MMP2 and 9, whereas females had significantly increased basal secretion of TNFα. This was caused by the smaller fragments of fetal DNA, whereas the larger fragments did not cause any significant increases. Male fetal DNA had significantly lower percentages of methylation than females. Thus, when the cytokine and pro-MMP activity data were correlated with methylation percentage, IL-6 secretion significantly correlated negatively, whereas GM-CSF secretion positively correlated. These data support the role of fetal DNA as an inflammatory stimulus in the FM, as measured by increased NF-κB translocation, cytokine secretion, and increased pro-MMP activity. However, the data also suggested that the responses are different from FM tissues of male and female fetuses, and both the fragment size and methylation status of the fetal DNA can influence the magnitude and type of molecule secreted.
Collapse
Affiliation(s)
- Chelsea A. Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | - Po’okela K. Ng
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | | | - Justin Padron
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Claire Enid Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- *Correspondence: Claire Enid Kendal-Wright,
| |
Collapse
|
17
|
Kurashima CK, Ng PK, Kendal-Wright CE. RAGE against the Machine: Can Increasing Our Understanding of RAGE Help Us to Battle SARS-CoV-2 Infection in Pregnancy? Int J Mol Sci 2022; 23:6359. [PMID: 35742804 PMCID: PMC9224312 DOI: 10.3390/ijms23126359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 12/05/2022] Open
Abstract
The receptor of advanced glycation end products (RAGE) is a receptor that is thought to be a key driver of inflammation in pregnancy, SARS-CoV-2, and also in the comorbidities that are known to aggravate these afflictions. In addition to this, vulnerable populations are particularly susceptible to the negative health outcomes when these afflictions are experienced in concert. RAGE binds a number of ligands produced by tissue damage and cellular stress, and its activation triggers the proinflammatory transcription factor Nuclear Factor Kappa B (NF-κB), with the subsequent generation of key proinflammatory cytokines. While this is important for fetal membrane weakening, RAGE is also activated at the end of pregnancy in the uterus, placenta, and cervix. The comorbidities of hypertension, cardiovascular disease, diabetes, and obesity are known to lead to poor pregnancy outcomes, and particularly in populations such as Native Hawaiians and Pacific Islanders. They have also been linked to RAGE activation when individuals are infected with SARS-CoV-2. Therefore, we propose that increasing our understanding of this receptor system will help us to understand how these various afflictions converge, how forms of RAGE could be used as a biomarker, and if its manipulation could be used to develop future therapeutic targets to help those at risk.
Collapse
Affiliation(s)
- Courtney K. Kurashima
- School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.K.K.); (P.K.N.)
| | - Po’okela K. Ng
- School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.K.K.); (P.K.N.)
| | - Claire E. Kendal-Wright
- School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.K.K.); (P.K.N.)
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai’i, Honolulu, HI 96813, USA
| |
Collapse
|
18
|
Menon R. Fetal inflammatory response at the fetomaternal interface: A requirement for labor at term and preterm. Immunol Rev 2022; 308:149-167. [PMID: 35285967 DOI: 10.1111/imr.13075] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022]
Abstract
Human parturition at term and preterm is an inflammatory process synchronously executed by both fetomaternal tissues to transition them from a quiescent state t an active state of labor to ensure delivery. The initiators of the inflammatory signaling mechanism can be both maternal and fetal. The placental (fetal)-maternal immune and endocrine mediated homeostatic imbalances and inflammation are well reported. However, the fetal inflammatory response (FIR) theories initiated by the fetal membranes (amniochorion) at the choriodecidual interface are not well established. Although immune cell migration, activation, and production of proparturition cytokines to the fetal membranes are reported, cellular level events that can generate a unique set of inflammation are not well discussed. This review discusses derangements to fetal membrane cells (physiologically and pathologically at term and preterm, respectively) in response to both endogenous and exogenous factors to generate inflammatory signals. In addition, the mechanisms of inflammatory signal propagation (fetal signaling of parturition) and how these signals cause immune imbalances at the choriodecidual interface are discussed. In addition to maternal inflammation, this review projects FIR as an additional mediator of inflammatory overload required to promote parturition.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
19
|
Short CES, Quinlan RA, Wang X, Preda VG, Smith A, Marchesi JR, Lee YS, MacIntyre DA, Bennett PR, Taylor GP. Vaginal Microbiota, Genital Inflammation and Extracellular Matrix Remodelling Collagenase: MMP-9 in Pregnant Women With HIV, a Potential Preterm Birth Mechanism Warranting Further Exploration. Front Cell Infect Microbiol 2021; 11:750103. [PMID: 34912728 PMCID: PMC8667959 DOI: 10.3389/fcimb.2021.750103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/03/2021] [Indexed: 01/24/2023] Open
Abstract
Background Pregnant women living with HIV infection (PWLWH) have elevated rates of preterm birth (PTB) in which HIV and cART are implicated. PWLWH also have a high prevalence of adverse vaginal microbiota, which associate with genital tract inflammation. The mechanism underlying PTB in PWLWH is unknown. We present the first data in PWLWH on genital-tract matrix-metalloproteinase-9(MMP-9), an important collagenase implicated in labour onset, and tissue inhibitor of metalloproteinases-1(TIMP-1) and explore correlations with local inflammation and vaginal bacteria. Material and Methods Cervical vaginal fluid (CVF) collected by a soft cup and high vaginal swabs (HVS) were obtained from PWLWH and HIV uninfected pregnant women (HUPW) at three antenatal time points. Maternal characteristics, combination antiretroviral therapy (cART) exposure, and pregnancy outcome were recorded. Concentrations of MMP-9, TIMP-1 and ten cytokines were measured by immunoassays. Vaginal microbiota composition was determined through 16S rRNA amplicon sequencing. MMP-9, TIMP-1 and cytokine concentrations were compared by HIV status, cART, and prematurity and in PWLWH correlations with polymorphonuclear leucocytes, cytokines and bacterial genera were explored. Results CVF was available for 50 PWLWH (108 samples) and 12 HUPW (20 samples) between gestation weeks 14-38. Thirty-six PWLWH conceived on cART and 14 initiated post-conception. There were five and one PTB outcomes in PWLWH and HUPW respectively. PWLWH had higher mean CVF concentrations of MMP-9 (p<0.001) and TIMP-1 (p=0.035) in the second trimester compared with HUPW with a similar trend in the third trimester. PWLWH also had higher CVF values of cytokines: IL-1β, IL-8, IL-12 and TNF-α in both trimesters compared to HUPW (p ≤ 0.003). In PWLWH, MMP-9 positively correlated with TIMP-1 (r=0.31, p=0.002) and CVF polymorphonuclear leucocytes (r=0.57, p=0.02). Correlations were observed between MMP-9 and three cytokines: IL-1β (r=0.61), IL-8 (r=0.57) and TNF-α (r=0.64), p<0.001, similarly for TIMP-1. Abundance of anaerobic pathobionts correlated with MMP-9: Gardnerella (r=0.44, p<0.001), Atopobium (r=0.33, p=0.005), and Prevotella genera (r=0.39, p<0.001). Conversely proportion of Lactobacillus genera negatively correlated with MMP-9 (rho=-0.46, p<0.001). MMP-9/TIMP-1 ratio increased with gestational age at sampling in PWLWH, but this was no longer significant after adjusting for confounders and no difference by prematurity was observed in this sub-study. Conclusions Here we show strong correlations of MMP-9 to genital tract inflammation and sub-optimal bacterial genera in PWLWH indicating the ascending genital tract infection pathway may be a contributory mechanism to the high risk of PTB.
Collapse
Affiliation(s)
- Charlotte-Eve S. Short
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Rachael A. Quinlan
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xuan Wang
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Veronica Georgiana Preda
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ann Smith
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Faculty of Health and Applied Sciences, University West of England, Bristol, United Kingdom
| | - Julian R. Marchesi
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Yooni S. Lee
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David A. MacIntyre
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Phillip R. Bennett
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Graham P. Taylor
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
- March of Dimes Prematurity Research Centre, Division of Development and Reproductive Biology, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Šket T, Ramuta TŽ, Starčič Erjavec M, Kreft ME. The Role of Innate Immune System in the Human Amniotic Membrane and Human Amniotic Fluid in Protection Against Intra-Amniotic Infections and Inflammation. Front Immunol 2021; 12:735324. [PMID: 34745106 PMCID: PMC8566738 DOI: 10.3389/fimmu.2021.735324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/23/2021] [Indexed: 01/18/2023] Open
Abstract
Intra-amniotic infection and inflammation (IAI) affect fetal development and are highly associated with preterm labor and premature rupture of membranes, which often lead to adverse neonatal outcomes. Human amniotic membrane (hAM), the inner part of the amnio-chorionic membrane, protects the embryo/fetus from environmental dangers, including microbial infection. However, weakened amnio-chorionic membrane may be breached or pathogens may enter through a different route, leading to IAI. The hAM and human amniotic fluid (hAF) respond by activation of all components of the innate immune system. This includes changes in 1) hAM structure, 2) presence of immune cells, 3) pattern recognition receptors, 4) cytokines, 5) antimicrobial peptides, 6) lipid derivatives, and 7) complement system. Herein we provide a comprehensive and integrative review of the current understanding of the innate immune response in the hAM and hAF, which will aid in design of novel studies that may lead to breakthroughs in how we perceive the IAI.
Collapse
Affiliation(s)
- Tina Šket
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Taja Železnik Ramuta
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
21
|
Choltus H, Lavergne M, De Sousa Do Outeiro C, Coste K, Belville C, Blanchon L, Sapin V. Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome. Biomedicines 2021; 9:biomedicines9091123. [PMID: 34572309 PMCID: PMC8466405 DOI: 10.3390/biomedicines9091123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/28/2022] Open
Abstract
Preterm prelabor ruptures of fetal membranes (pPROM) are a pregnancy complication responsible for 30% of all preterm births. This pathology currently appears more as a consequence of early and uncontrolled process runaway activation, which is usually implicated in the physiologic rupture at term: inflammation. This phenomenon can be septic but also sterile. In this latter case, the inflammation depends on some specific molecules called “alarmins” or “damage-associated molecular patterns” (DAMPs) that are recognized by pattern recognition receptors (PRRs), leading to a microbial-free inflammatory response. Recent data clarify how this activation works and which receptor translates this inflammatory signaling into fetal membranes (FM) to manage a successful rupture after 37 weeks of gestation. In this context, this review focused on two PRRs: the receptor for advanced glycation end-products (RAGE) and the NLRP7 inflammasome.
Collapse
Affiliation(s)
- Helena Choltus
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Marilyne Lavergne
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Coraline De Sousa Do Outeiro
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Karen Coste
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Corinne Belville
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Loïc Blanchon
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Vincent Sapin
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
- CHU de Clermont-Ferrand, Biochemistry and Molecular Genetic Department, 63000 Clermont-Ferrand, France
- Correspondence: ; Tel.: +33-473-178-174
| |
Collapse
|
22
|
Amberg BJ, Hodges RJ, Rodgers KA, Crossley KJ, Hooper SB, DeKoninck PLJ. Why Do the Fetal Membranes Rupture Early after Fetoscopy? A Review. Fetal Diagn Ther 2021; 48:493-503. [PMID: 34404043 DOI: 10.1159/000517151] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/03/2021] [Indexed: 11/19/2022]
Abstract
Iatrogenic preterm premature rupture of the fetal membranes (iPPROM) remains the Achilles' heel of keyhole fetal surgery (fetoscopy) despite significant efforts in preclinical models to develop new therapies. This limited success is partially due to incomplete understanding why the fetal membranes rupture early after fetoscopy and notable differences in membrane physiology between humans and domestic species. In this review, we summarize aspects of fetoscopy that may contribute to iPPROM, the previous efforts to develop new therapies, and limitations of preclinical models commonly used in fetal membrane research.
Collapse
Affiliation(s)
- Benjamin J Amberg
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia, .,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia,
| | - Ryan J Hodges
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Karyn A Rodgers
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Kelly J Crossley
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Stuart B Hooper
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Philip L J DeKoninck
- The Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
23
|
Saito Reis CA, Padron JG, Norman Ing ND, Kendal-Wright CE. High-mobility group box 1 is a driver of inflammation throughout pregnancy. Am J Reprod Immunol 2020; 85:e13328. [PMID: 32851715 DOI: 10.1111/aji.13328] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
A proinflammatory response driven by high-mobility group box 1 (HMGB1) is important for the success of both the early stages of pregnancy and parturition initiation. However, the tight regulation of HMGB1 within these two stages is critical, as increased HMGB1 can manifest into pregnancy-related pathologies. Although during the early stages of pregnancy HMGB1 is critical for the development and implantation of the embryo, and uterine decidualization, high levels within the uterine cavity have been linked to pregnancy failure. In addition, chronic inflammation, resultant from increased HMGB1 within the maternal circulation and gestational tissues, also increases the risk for preterm labor, preterm birth, or infant mortality. Due to the link between HMGB1 and several pregnancy pathologies, the possibility of leveraging HMGB1 as a biomarker has been assessed. However, data are limited that demonstrate how known HMGB1 inhibitors could reduce inflammation within pregnancy. Thus, further research is warranted to improve our understanding of the potential of HMGB1 as a therapeutic target to reduce inflammation within pregnancy. This review aims to describe what is understood about the role of HMGB1 that drives inflammation throughout pregnancy and highlight its potential as a biomarker and therapeutic target within this context.
Collapse
Affiliation(s)
- Chelsea A Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Justin G Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA
| | - Nainoa D Norman Ing
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Claire E Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA.,Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA.,Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawai'I at Manoā, Honolulu, HI, USA
| |
Collapse
|
24
|
Menon R, Behnia F, Polettini J, Richardson LS. Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm pre-labor rupture of the membranes. Semin Immunopathol 2020; 42:431-450. [PMID: 32785751 DOI: 10.1007/s00281-020-00808-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Spontaneous preterm birth (PTB) and preterm pre-labor rupture of the membranes (pPROM) are major pregnancy complications. Although PTB and pPROM have common etiologies, they arise from distinct pathophysiologic pathways. Inflammation is a common underlying mechanism in both conditions. Balanced inflammation is required for fetoplacental growth; however, overwhelming inflammation (physiologic at term and pathologic at preterm) can lead to term and preterm parturition. A lack of effective strategies to control inflammation and reduce the risk of PTB and pPROM suggests that there are several modes of the generation of inflammation which may be dependent on the type of uterine tissue. The avascular fetal membrane (amniochorion), which provides structure, support, and protection to the intrauterine cavity, is one of the key contributors of inflammation. Localized membrane inflammation helps tissue remodeling during pregnancy. Two unique mechanisms that generate balanced inflammation are the progressive development of senescence (aging) and cyclic cellular transitions: epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET). The intrauterine build-up of oxidative stress at term or in response to risk factors (preterm) can accelerate senescence and promote a terminal state of EMT, resulting in the accumulation of inflammation. Inflammation degrades the matrix and destabilizes membrane function. Inflammatory mediators from damaged membranes are propagated via extracellular vesicles (EV) to maternal uterine tissues and transition quiescent maternal uterine tissues into an active state of labor. Membrane inflammation and its propagation are fetal signals that may promote parturition. This review summarizes the mechanisms of fetal membrane cellular senescence, transitions, and the generation of inflammation that contributes to term and preterm parturitions.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA.
| | - Faranak Behnia
- Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, UT Health, Houston, Texas, USA
| | - Jossimara Polettini
- Universidade Federal da Fronteira Sul, Campus Passo Fundo, Rua Capitão Araujo, 20, Centro, Passo Fundo, Rio Grande do Sul, Brazil
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA
| |
Collapse
|