1
|
Starodubtseva N, Tokareva A, Kononikhin A, Brzhozovskiy A, Bugrova A, Kukaev E, Muminova K, Nakhabina A, Frankevich VE, Nikolaev E, Sukhikh G. First-Trimester Preeclampsia-Induced Disturbance in Maternal Blood Serum Proteome: A Pilot Study. Int J Mol Sci 2024; 25:10653. [PMID: 39408980 PMCID: PMC11476624 DOI: 10.3390/ijms251910653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Preeclampsia (PE) is a complex and multifaceted obstetric syndrome characterized by several distinct molecular subtypes. It complicates up to 5% of pregnancies and significantly contributes to maternal and newborn morbidity, thereby diminishing the long-term quality of life for affected women. Due to the widespread dissatisfaction with the effectiveness of existing approaches for assessing PE risk, there is a pressing need for ongoing research to identify newer, more accurate predictors. This study aimed to investigate early changes in the maternal serum proteome and associated signaling pathways. The levels of 125 maternal serum proteins at 11-13 weeks of gestation were quantified using liquid chromatography-multiple reaction monitoring mass spectrometry (LC-MRM MS) with the BAK-125 kit. Ten serum proteins emerged as potential early markers for PE: Apolipoprotein M (APOM), Complement C1q subcomponent subunit B (C1QB), Lysozyme (LYZ), Prothrombin (F2), Albumin (ALB), Zinc-alpha-2-glycoprotein (AZGP1), Tenascin-X (TNXB), Alpha-1-antitrypsin (SERPINA1), Attractin (ATRN), and Apolipoprotein A-IV (APOA4). Notably, nine of these proteins have previously been associated with PE in prior research, underscoring the consistency and reliability of our findings. These proteins play key roles in critical molecular processes, including complement and coagulation cascades, platelet activation, and insulin-like growth factor pathways. To improve the early prediction of PE, a highly effective Support Vector Machine (SVM) model was developed, analyzing 19 maternal serum proteins from the first trimester. This model achieved an area under the curve (AUC) of 0.91, with 87% sensitivity and 95% specificity, and a hazard ratio (HR) of 13.5 (4.6-40.8) with p < 0.001. These findings demonstrate that serum protein-based SVM models possess significantly higher predictive power compared to the routine first-trimester screening test, highlighting their superior utility in the early detection and risk stratification of PE.
Collapse
Affiliation(s)
- Natalia Starodubtseva
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
- Moscow Center for Advanced Studies, 123592 Moscow, Russia
| | - Alisa Tokareva
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| | - Alexey Kononikhin
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| | - Alexander Brzhozovskiy
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| | - Anna Bugrova
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Evgenii Kukaev
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
- V.L. Talrose Institute for Energy Problems of Chemical Physics, N.N. Semenov Federal Research Center of Chemical Physics, 119334 Moscow, Russia
| | - Kamilla Muminova
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| | - Alina Nakhabina
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| | - Vladimir E. Frankevich
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
- Laboratory of Translational Medicine, Siberian State Medical University, 634050 Tomsk, Russia
| | | | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics Gynecology and Perinatology, Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia; (A.T.); (A.K.); (A.B.); (A.B.); (E.K.); (K.M.); (A.N.); (V.E.F.); (G.S.)
| |
Collapse
|
2
|
Ali M, Ahmed M, Memon M, Chandio F, Shaikh Q, Parveen A, Phull AR. Preeclampsia: A comprehensive review. Clin Chim Acta 2024; 563:119922. [PMID: 39142550 DOI: 10.1016/j.cca.2024.119922] [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: 07/03/2024] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Preeclampsia (PE) is a life-threatening disease of pregnancy and a prominent cause of neonatal and maternal mortality and morbidity. PE affects approximately 5-10% of pregnancies worldwide, posing significant risks to perinatal and maternal health. It is characterized by a variety of interconnected pathological cascades contributing to the stimulation of intravascular inflammation, oxidative stress (OS), endothelial cell activation, and syncytiotrophoblast stress that converge on a common pathway, ultimately resulting in disease progression. The present study was designed and executed to review the existing scientific literature, specifically focusing on the etiology (gestational diabetes mellitus and maternal obesity, insulin resistance, metabolic syndrome, maternal infection, periodontal disease, altered microbiome, and genetics), clinical presentations (hypertension, blood disorders, proteinuria, hepatic dysfunction, renal dysfunction, pulmonary edema, cardiac dysfunction, fetal growth restrictions, and eclampsia), therapeutic clinical biomarkers (creatinine, albuminuria, and cystatin C) along with their associations and mechanisms in PE. In addition, this study provides insights into the potential of nanomedicines for targeting these mechanisms for PE management and treatment. Inflammation, OS, proteinuria, and an altered microbiome are prominent biomarkers associated with progression and PE-related pathogenesis. Understanding the molecular mechanisms, exploring suitable markers, targeted interventions, comprehensive screening, and holistic strategies are critical to decreasing the incidence of PE and promoting maternal-fetal well-being. The present study comprehensively reviewed the etiology, clinical presentations, therapeutic biomarkers, and preventive potential of nanomedicines in the treatment and management of PE.
Collapse
Affiliation(s)
- Majida Ali
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Madiha Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Jaffer Khan Jamali Road, H-8/4, Islamabad, Pakistan
| | - Mehwish Memon
- Department of Biochemistry, Ibn e Sina University, Mirpur Khas, Pakistan
| | - Fozia Chandio
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Quratulain Shaikh
- Department of Gynecology and Obstetrics, Shaikh Zaid Women Hospital Larkana, Shaheed Mohtarma Benazir Bhutto Medical University (SMBB) Larkana, Pakistan
| | - Amna Parveen
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, South Korea.
| | - Abdul-Rehman Phull
- Department of Biochemistry, Shah Abdul Latif University, Khairpur, Sindh, Pakistan.
| |
Collapse
|
3
|
Ramdin S, Naicker T, Baijnath S, Govender N. Is renal dysfunction amplified in an arginine vasopressin induced rat model of preeclampsia? Reprod Biol 2024; 24:100910. [PMID: 38851025 DOI: 10.1016/j.repbio.2024.100910] [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: 09/19/2023] [Revised: 04/27/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Renal dysfunction is important in preeclampsia (PE) pathophysiology and has not been fully explored in the arginine vasopressin (AVP) rat model of PE. This study aimed to determine kidney toxicity associated with this model. Female Sprague Dawley rats (n = 24) were subcutaneously infused with AVP or saline for 18 days. Urine samples (GD8, 14 and 18) were used to determine the levels of albumin, VEGF-A, clusterin, NGAL/Lipocalin-2, KIM-1, cystatin C, TIMP-1, β2M and OPN via Multiplex ELISAs. Albumin, and NGAL/lipocalin-2 were significantly elevated in the PAVP vs PS group on GD14 and GD18 (p < 0.001) respectively. VEGF-A significantly decreased in the pregnant vs non-pregnant groups on GD14 and 18 (p < 0.001). Clusterin (p < 0.001) and OPN (p < 0.05) were significantly higher in the PAVP vs PS group on GD18. Cystatin C and KIM-1 are significantly upregulated in the PAVP vs PS groups throughout gestation (p < 0.05). β2M is significantly elevated in the PAVP vs PS group on GD14 and 18 (p < 0.05). AVP elevated the urinary levels of the kidney injury biomarkers and replicated the renal dysfunction associated with PE development. Our findings confirm the potential applications of this model in studying the mechanisms underlying renal damage in PE.
Collapse
Affiliation(s)
- Sapna Ramdin
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sooraj Baijnath
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nalini Govender
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa.
| |
Collapse
|
4
|
Fast MS, Weyer K, Pedersen H, Andersen GR, Birn H. Filtration and tubular handling of EWE-hC3Nb1, a complement inhibitor nanobody, in wild type mice and a mouse model of proteinuric kidney disease. FEBS Open Bio 2024; 14:322-330. [PMID: 38124617 PMCID: PMC10839346 DOI: 10.1002/2211-5463.13752] [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: 10/17/2023] [Revised: 11/26/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Tubular activation and deposition of filtered complement proteins have been implicated in the progression of proteinuric kidney disease. The potent C3b-specific nanobody inhibitor of the alternative pathway, EWE-hC3Nb1, is likely freely filtered in the glomerulus to allow complement inhibition in the tubular lumen and may provide a novel treatment option to prevent tubulointerstitial injury. However, more information on the pharmacokinetic properties and renal tubular handling of EWE-hC3Nb1 nanobody is required for its pharmacological application in relation to kidney disease. Here, we examined the pharmacokinetic properties of free EWE-hC3Nb1 in mouse plasma and urine, following subcutaneous injection in wild-type control and podocin knock out (KO) mice with severe proteinuria. Tubular handling of filtered EWE-hC3Nb1 was assessed by immunohistochemistry (IHC) on kidney tissue from control, proteinuric mice, and KO mice deficient in the proximal tubule endocytic receptor megalin. Rapid plasma absorption and elimination of EWE-hC3Nb1 was observed in both control and proteinuric mice; however, urinary excretion of EWE-hC3Nb1 was markedly increased in proteinuric mice. Urinary EWE-hC3Nb1 excretion was amplified in megalin KO mice, and substantial accumulation of EWE-hC3Nb1 was observed in megalin-expressing renal proximal tubules by IHC. Moreover, free EWE-hC3Nb1 was found to be rapidly cleared from plasma. In conclusion, filtered EWE-hC3Nb1 is reabsorbed by a megalin-dependent process in the proximal tubules. Increased load of filtered proteins in the tubular fluid may inhibit the megalin-dependent uptake of EWE-hC3Nb1 in proteinuric mice. Treatment with EWE-hC3Nb1 may allow investigation of the effects of complement inhibition in the tubular fluid.
Collapse
Affiliation(s)
| | | | - Henrik Pedersen
- Department of Molecular Biology and Genetics – Protein ScienceAarhus UniversityDenmark
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics – Protein ScienceAarhus UniversityDenmark
| | - Henrik Birn
- Department of BiomedicineAarhus UniversityDenmark
- Departments of Clinical MedicineAarhus University and Renal Medicine, Aarhus University HospitalDenmark
| |
Collapse
|
5
|
Avendanha RA, Campos GFC, Branco BC, Ishii NC, Gomes LHN, de Castro AJ, Leal CRV, Simões E Silva AC. Potential urinary biomarkers in preeclampsia: a narrative review. Mol Biol Rep 2024; 51:172. [PMID: 38252179 DOI: 10.1007/s11033-023-09053-5] [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: 05/03/2023] [Accepted: 10/30/2023] [Indexed: 01/23/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a highly relevant pregnancy-related disorder. An early and accurate diagnosis is crucial to prevent major maternal and neonatal complications and mortality. Due to the association of kidney dysfunction with the pathophysiology of the disease, urine samples have the potential to provide biomarkers for PE prediction, being minimally invasive and easy to perform. Therefore, searching for novel biomarkers may improve outcomes. This narrative review aimed to summarize the scientific literature about the traditional and potential urinary biomarkers in PE and to investigate their applicability to screen and diagnose the disorder. METHODS A non-systematic search was performed in PubMed/MEDLINE, Scopus, and SciELO databases. RESULTS There is significant divergence in the literature regarding traditionally used serum markers creatinine, cystatin C, and albuminuria, accuracy in PE prediction. As for the potential renal biomarkers investigated, including vascular epithelial growth factor (VEGF), placental growth factor (PlGF), and soluble fms-like tyrosine kinase (sFlt-1), urinary levels of PlGF and sFtl-1/PlGF ratio in urine seem to be the most promising as screening tests. The assessment of the global load of misfolded proteins through urinary congophilia, podocyturia, and nephrinuria has also shown potential for screening and diagnosis. Studies regarding the use of proteomics and metabolomics have shown good accuracy, sensitivity, and specificity for predicting the development and severity of PE. CONCLUSION However, there are still many divergences in the literature, which requires future and more conclusive research to confirm the predictive role of urinary biomarkers in pregnant women with PE.
Collapse
Affiliation(s)
- Renata Araujo Avendanha
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Beatriz Castello Branco
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil
| | - Nicolle Coimbra Ishii
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Universidade Federal de Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - Luiz Henrique Nacife Gomes
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Faculdade de Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ailton José de Castro
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Caio Ribeiro Vieira Leal
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Cristina Simões E Silva
- Liga Acadêmica de Pesquisa Científica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, UFMG, Belo Horizonte, MG, Brazil.
- Faculdade de Medicina, UFMG, Avenida Alfredo Balena, 190, 2o andar, sala 281. Bairro Santa Efigênia, Belo Horizonte, CEP 30130-100, MG, Brazil.
| |
Collapse
|
6
|
Wang X, Shields CA, Ekperikpe U, Amaral LM, Williams JM, Cornelius DC. VASCULAR AND RENAL MECHANISMS OF PREECLAMPSIA. CURRENT OPINION IN PHYSIOLOGY 2023; 33:100655. [PMID: 37009057 PMCID: PMC10062189 DOI: 10.1016/j.cophys.2023.100655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Preeclampsia (PE) is a multisystem obstetric disorder that affects 2-10% of pregnancies worldwide and it is a leading cause of maternal and fetal morbidity and mortality. The etiology of PE development is not clearly delineated, but since delivery of the fetus and placenta often leads to symptom resolution in the most cases of PE, it is hypothesized that the placenta is the inciting factor of the disease. Current management strategies for PE focus on treating the maternal symptoms to stabilize the mother in an attempt to prolong the pregnancy. However, the efficacy of this management strategy is limited. Therefore, identification of novel therapeutic targets and strategies is needed. Here, we provide a comprehensive overview of the current state of knowledge regarding mechanisms of vascular and renal pathophysiology during PE and discuss potential therapeutic targets directed at improving maternal vascular and renal function.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology, University of Mississippi Medical Center
| | - Corbin A Shields
- Department of Emergency Medicine, University of Mississippi Medical Center
| | - Ubong Ekperikpe
- Department of Pharmacology, University of Mississippi Medical Center
| | - Lorena M Amaral
- Department of Pharmacology, University of Mississippi Medical Center
| | | | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center
- Department of Emergency Medicine, University of Mississippi Medical Center
| |
Collapse
|
7
|
Isaksson GL, Nielsen LH, Palarasah Y, Jensen DM, Andersen LLT, Madsen K, Bistrup C, Jørgensen JS, Ovesen PG, Jensen BL. Urine excretion of C3dg and sC5b-9 coincide with proteinuria and development of preeclampsia in pregnant women with type-1 diabetes. J Hypertens 2023; 41:223-232. [PMID: 36583350 DOI: 10.1097/hjh.0000000000003288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Pregnant women with type-1 diabetes have an increased risk of preeclampsia with kidney injury and cardiovascular complications. Urine excretion of plasmin and soluble membrane attack complex (sC5b-9) is elevated in severe preeclampsia. We hypothesized a coupling between these events and that active plasmin promotes intratubular complement activation and membrane deposition. METHODS Stored urine and plasma samples from pregnant women with type-1 diabetes (n = 88) collected at gestational weeks 12, 20, 28, 32, 36 and 38 were used. In the cohort, 14 women developed preeclampsia and were compared with 16 nonpreeclampsia controls. RESULTS Urine C3dg and sC5b-9-associated C9 neoantigen/creatinine ratios increased and were significantly higher in women who developed preeclampsia. Plasma concentrations did not change with gestation. Urine plasmin(ogen) correlated to urine C3dg (r = 0.51, P < 0.001) and C9 neoantigen (r = 0.68, P < 0.001); urine albumin correlated to C3dg (r = 0.44, P < 0.001) and C9 (r = 0.59, P < 0.001). Membrane-associated C3dg and C9 neoantigen was detected in urinary extracellular vesicles from patients but not controls at 36 weeks. Receiver operating characteristic curves showed that C3dg and C9 neoantigen were inferior to albumin as predictive biomarkers for preeclampsia. CONCLUSION In preeclampsia, urinary excretion of activated complement relates significantly to albuminuria and to plasmin(ogen) but not to activation in plasma. Intratubular complement activation in preeclampsia is a postfiltration event tightly related to proteinuria/plasminogenuria and a possible mechanistic link to cellular damage and kidney injury.
Collapse
Affiliation(s)
- Gustaf L Isaksson
- Department of Nephrology, Odense University Hospital, Odense
- Department of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark
| | - Lise H Nielsen
- Department of Clinical Medicine - Obstetrics and Gynecology, Aarhus University, Aarhus
| | - Yaseelan Palarasah
- Department of Molecular Medicine - Cancer and Inflammation, University of Southern Denmark
| | - Dorte M Jensen
- Steno Diabetes Center Odense, Odense University Hospital
- Department of Clinical Research, University of Southern Denmark
| | - Lise L T Andersen
- Department of Clinical Research, University of Southern Denmark
- Department of Obstetrics and Gynecology, Odense University Hospital
| | - Kirsten Madsen
- Department of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Claus Bistrup
- Department of Nephrology, Odense University Hospital, Odense
- Department of Clinical Research, University of Southern Denmark
| | - Jan S Jørgensen
- Department of Clinical Research, University of Southern Denmark
- Department of Obstetrics and Gynecology, Odense University Hospital
| | - Per G Ovesen
- Department of Clinical Medicine - Obstetrics and Gynecology, Aarhus University, Aarhus
| | - Boye L Jensen
- Department of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark
| |
Collapse
|
8
|
Complement activation and regulation in preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome. Am J Obstet Gynecol 2022; 226:S1059-S1070. [PMID: 32986992 DOI: 10.1016/j.ajog.2020.09.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022]
Abstract
The complement system is critical to human health owing to its central role in host defense and innate immunity. During pregnancy, the complement system must be appropriately regulated to allow for immunologic tolerance to the developing fetus and placenta. Although some degree of complement activation can be seen in normal pregnancy, the fetus seems to be protected in part through the placental expression of complement regulatory proteins, which inhibit complement activation at different steps along the complement activation cascade. In women who develop preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, there is a shift toward increased complement activation and decreased complement regulation. There is an increase in placental deposition of C5b-9, which is the terminal effector of classical, lectin, and alternative complement pathways. C5b-9 deposition stimulates trophoblasts to secrete soluble fms-like tyrosine kinase-1, which sequesters vascular endothelial growth factor and placental growth factor. Pathogenic mutations or deletions in complement regulatory genes, which predispose to increased complement activation, have been detected in women with preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome. Before the disease, biomarkers of alternative complement pathway activation are increased; during active disease, biomarkers of terminal complement pathway activation are increased. Urinary excretion of C5b-9 is associated with preeclampsia with severe features and distinguishes it from other hypertensive disorders of pregnancy. Taken together, existing data link preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome with increased activation of the terminal complement pathway that, in some cases, may be influenced by genetic alterations in complement regulators. These findings suggest that the inhibition of the terminal complement pathway, possibly through C5 blockade, may be an effective strategy to treat preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, but this strategy warrants further evaluation in clinical trials.
Collapse
|
9
|
Berenji MG, Berenji HG, Pashapour S, Sadeghpour S. Serum Netrin-1 and Urinary KIM-1 levels as potential biomarkers for the diagnosis of early preeclampsia. J OBSTET GYNAECOL 2021; 42:636-640. [PMID: 34569430 DOI: 10.1080/01443615.2021.1945010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The aim of this study was to evaluate whether the Serum Netrin-1 and Urinary KIM-1 (Kidney Injury Molecule-1) levels are associated with the detection of preeclampsia. A total of 90 patients, including 36 normal pregnant women, 29 patients with nonsevere preeclampsia and 25 patients with severe preeclampsia, were included in this study. Maternal serum Netrin-1 and Urinary KIM-1 levels were measured by using an enzyme-linked immunosorbent assay (ELISA). The results showed that the Levels of Netrin-1 and KIM-1 were statistically higher in women with preeclampsia as compared with normal pregnant women. Furthermore, the Netrin-1 level in women with severe preeclampsia was significantly higher than nonsevere preeclamptic women. inconclusion the current study showed that Maternal serum level of Netrin-1 and Urinary level of KIM-1 can be used as early biomarkers for the detection of preeclampsia.IMPACT STATEMENTWhat is already known on this subject? Preeclampsia is a disorder of widespread vascular endothelial malfunction and vasospasm that occurs after 20 weeks' gestation. Netrin-1 was found to promote angiogenesis. Alteration of placental angiogenesis in early pregnancy is a well-known reason for placental dysfunction such as preeclampsia. Kidney injury with proteinuria is a characteristic feature of preeclampsia. Urine KIM-1 is the most potential biomarker for renal injury in preeclampsia. Due to these facts, we aimed to investigate the role of maternal serum Netrin-1 and Urine KIM-1 levels in preeclampsia presence and severity.What the results of this study add? A significant relationship between Netrin-1 and KIM-1 levels with preeclampsia.What the implications are of these findings for clinical practice and/or further research? Based on these findings, we concluded that increased levels of Netrin-1 and KIM-1 are associated with severe preeclampsia.
Collapse
Affiliation(s)
- Morteza Ghasemnejad Berenji
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Hojjat Ghasemnejad Berenji
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarvin Pashapour
- Department of Pediatrics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sonia Sadeghpour
- Department of Obstetrics and Gynecology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
10
|
Effect of preeclampsia and its severity on maternal serum NGAL and KIM-1 levels during pregnancy and the post-pregnancy period. Eur J Obstet Gynecol Reprod Biol 2020; 256:246-251. [PMID: 33248380 DOI: 10.1016/j.ejogrb.2020.11.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/07/2020] [Accepted: 11/12/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The purpose of this trial was to appraise the effects of preeclampsia and its intensity on maternal serum neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) levels during pregnancy and the post-pregnancy period. STUDY DESIGN Firstly pregnant participants (n = 156) were separated into three groups, as control, mild, and severe preeclampsia. Secondly women in post-pregnancy period (n = 368) were separated into three groups according to history of pregnancy, as healthy control, mild, and severe preeclampsia. These women were identified through the hospital data system and contacted by telephone to participate in the study. RESULTS Our study comprised 147 patients, 77 of whom were pregnant and 70 of whom were in their post-pregnancy period after the exclusion criteria had been applied. In terms of maternal serum NGAL levels, there is a significant increase in the severe preeclampsia group compared with that in the mild preeclampsia and normal pregnancy groups (p < 0.001). During the post-pregnancy period, the maternal serum NGAL levels were found significantly higher in the severe preeclampsia group than in the mild preeclampsia group and non-hypertension control group (p < 0.001). Maternal serum KIM-1 levels were found as significantly higher in the severe and mild preeclampsia groups than in the non-hypertension pregnancy group (p = 0.004). During the post-pregnancy period, maternal serum KIM-1 levels were found as similar among all post pregnant groups (p = 0.792). CONCLUSIONS Our results indicated that as the severity of preeclampsia increases, kidney damage as assessed using NGAL levels continues for a long period of time, even during the post-pregnancy period.
Collapse
|
11
|
Barnum SR, Bubeck D, Schein TN. Soluble Membrane Attack Complex: Biochemistry and Immunobiology. Front Immunol 2020; 11:585108. [PMID: 33240274 PMCID: PMC7683570 DOI: 10.3389/fimmu.2020.585108] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
The soluble membrane attack complex (sMAC, a.k.a., sC5b-9 or TCC) is generated on activation of complement and contains the complement proteins C5b, C6, C7, C8, C9 together with the regulatory proteins clusterin and/or vitronectin. sMAC is a member of the MACPF/cholesterol-dependent-cytolysin superfamily of pore-forming molecules that insert into lipid bilayers and disrupt cellular integrity and function. sMAC is a unique complement activation macromolecule as it is comprised of several different subunits. To date no complement-mediated function has been identified for sMAC. sMAC is present in blood and other body fluids under homeostatic conditions and there is abundant evidence documenting changes in sMAC levels during infection, autoimmune disease and trauma. Despite decades of scientific interest in sMAC, the mechanisms regulating its formation in healthy individuals and its biological functions in both health and disease remain poorly understood. Here, we review the structural differences between sMAC and its membrane counterpart, MAC, and examine sMAC immunobiology with respect to its presence in body fluids in health and disease. Finally, we discuss the diagnostic potential of sMAC for diagnostic and prognostic applications and potential utility as a companion diagnostic.
Collapse
Affiliation(s)
| | - Doryen Bubeck
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
12
|
Late first trimester circulating microparticle proteins predict the risk of preeclampsia < 35 weeks and suggest phenotypic differences among affected cases. Sci Rep 2020; 10:17353. [PMID: 33087742 PMCID: PMC7578826 DOI: 10.1038/s41598-020-74078-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
We hypothesize that first trimester circulating micro particle (CMP) proteins will define preeclampsia risk while identifying clusters of disease subtypes among cases. We performed a nested case–control analysis among women with and without preeclampsia. Cases diagnosed < 34 weeks’ gestation were matched to controls. Plasma CMPs were isolated via size exclusion chromatography and analyzed using global proteome profiling based on HRAM mass spectrometry. Logistic models then determined feature selection with best performing models determined by cross-validation. K-means clustering examined cases for phenotypic subtypes and biological pathway enrichment was examined. Our results indicated that the proteins distinguishing cases from controls were enriched in biological pathways involved in blood coagulation, hemostasis and tissue repair. A panel consisting of C1RL, GP1BA, VTNC, and ZA2G demonstrated the best distinguishing performance (AUC of 0.79). Among the cases of preeclampsia, two phenotypic sub clusters distinguished cases; one enriched for platelet degranulation and blood coagulation pathways and the other for complement and immune response-associated pathways (corrected p < 0.001). Significantly, the second of the two clusters demonstrated lower gestational age at delivery (p = 0.049), increased protein excretion (p = 0.01), more extreme laboratory derangement (p < 0.0001) and marginally increased diastolic pressure (p = 0.09). We conclude that CMP-associated proteins at 12 weeks’ gestation predict the overall risk of developing early preeclampsia and indicate distinct subtypes of pathophysiology and clinical morbidity.
Collapse
|
13
|
Plasma CD59 concentrations are increased in preeclampsia with severe features and correlate with laboratory measures of end-organ injury. Pregnancy Hypertens 2020; 22:204-209. [PMID: 33091682 DOI: 10.1016/j.preghy.2020.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/08/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Dysregulation of CD59 may lead to increased complement-mediated end-organ injury in preeclampsia. We sought to determine if soluble CD59 concentrations are altered in preeclampsia with severe features. STUDY DESIGN Observational case-control study, which enrolled subjects prospectively from six centers in Colombia from 2015 to 2016. Cases had preeclampsia with severe features and controls were either healthy or had chronic hypertension, gestational hypertension, or preeclampsia without severe features. Trained coordinators collected clinical data, blood and urine. Analyses were by test of medians and Spearman's correlation. MAIN OUTCOME MEASURES Soluble CD59 concentration in plasma and urine, using enzyme linked immunosorbent assays. RESULTS In total, 352 subjects were enrolled (104 cases; 248 controls). Compared to healthy women or those with other hypertensive disorders of pregnancy, women with preeclampsia with severe features had increased concentration of CD59 in plasma (P < 0.001) and decreased CD59 in urine (P = 0.01). In sub-group analyses, plasma CD59 concentrations were increased in preeclampsia with severe features compared to healthy controls (P < 0.001) or controls with either chronic hypertension (P = 0.002) or gestational hypertension (P = 0.02). Increased plasma CD59 concentrations correlated with decreased platelet count and increased lactate dehydrogenase, creatinine, aspartate transaminase, urine protein/creatinine ratio, systolic blood pressure and diastolic blood pressure (P < 0.01, all correlations). CONCLUSION In women with preeclampsia with severe features, soluble CD59 concentrations were increased in plasma and decreased in urine, and plasma levels correlated with increased blood pressure and end-organ injury. Soluble CD59 concentrations may help identify a subset of women with preeclampsia that have altered regulation of terminal complement proteins.
Collapse
|
14
|
Albejante MC, Kunz TCM, Ferreira MFC, Júnior JHZR, de Almeida RJ, Bacigalupo LDS, Matheus LHG, Dalboni MA, Camacho CP, Dellê H. Proteinuria is Associated with Urinary Loss of Cubilin and Vitamin D-Binding Protein in Patients with Preeclampsia. Sci Rep 2020; 10:3956. [PMID: 32127613 PMCID: PMC7054416 DOI: 10.1038/s41598-020-60924-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/10/2020] [Indexed: 11/09/2022] Open
Abstract
Women with preeclampsia (PE) form a vulnerable group for vitamin D3 deficiency. Reabsorption of vitamin D3 occurs in the proximal tubule after being endocytosed in combination with DBP (vitamin D binding protein) by the megalin/cubilin receptor. Because proteinuria promotes tubule injury and dysfunction, we hypothesized that the proteinuria present in PE could promote the loss of these components into the urine. Twenty preeclamptic patients and ten normal pregnant women with a gestational age greater than 20 weeks composed three groups: NC, normotensive control pregnant patients; PE, non-proteinuric preeclamptic patients; and PPE, preeclamptic patients with proteinuria. When proteinuria was absent, preeclampsia was diagnosed accordingly to the American College of Obstetricians and Gynecologists' (ACOG) guideline. The presence of 24-hour proteinuria equal to or greater than 300 mg was considered to form the PPE group. Urinary cubilin, megalin, and DBP were measured by ELISA and normalized by urinary creatinine. Regarding gestational age, there was no difference between the groups. NC group had arterial pressure within normal values, whereas PE and PPE groups had a significant increase (p < 0.01). As expected, PPE group presented elevated ACR (p < 0.05), accompanied by large amounts of cubilin and DBP in the urine (p < 0.05 vs. NC and PE). No difference was found in urinary megalin. PPE patients showed more chance of shedding cubilin into the urine compared to non-proteinuric patients (odds ratio 12.7 (1.2-136.3). In conclusion, this study further tightens the relationship between PE and vitamin D3 deficiency, since proteinuria present in PE induces the loss of molecules responsible for renal tubular vitamin D3 reabsorption for subsequent activation. Combined with other factors, the proteinuria may intensify vitamin D3 deficiency in PE.
Collapse
Affiliation(s)
- Maria Clara Albejante
- Postgraduate Program in Medicine, Universidade Nove de Julho - UNINOVE, São Paulo, Brazil
| | | | | | | | - Robson José de Almeida
- Postgraduate Program in Medicine, Universidade Nove de Julho - UNINOVE, São Paulo, Brazil
| | | | | | | | - Cleber Pinto Camacho
- Postgraduate Program in Medicine, Universidade Nove de Julho - UNINOVE, São Paulo, Brazil
| | - Humberto Dellê
- Postgraduate Program in Medicine, Universidade Nove de Julho - UNINOVE, São Paulo, Brazil.
| |
Collapse
|
15
|
Santelli A, Sun IO, Eirin A, Abumoawad AM, Woollard JR, Lerman A, Textor S, Puranik AS, Lerman LO. Senescent Kidney Cells in Hypertensive Patients Release Urinary Extracellular Vesicles. J Am Heart Assoc 2019; 8:e012584. [PMID: 31433703 PMCID: PMC6585370 DOI: 10.1161/jaha.119.012584] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
Abstract
Background Hypertension may be associated with renal cellular injury. Cells in distress release extracellular vesicles (EVs), and their numbers in urine may reflect renal injury. Cellular senescence, an irreversible growth arrest in response to a noxious milieu, is characterized by release of proinflammatory cytokines. We hypothesized that EVs released by senescent nephron cells can be identified in urine of patients with hypertension. Methods and Results We recruited patients with essential hypertension (EH) or renovascular hypertension and healthy volunteers (n=14 each). Renal oxygenation was assessed using magnetic resonance imaging and blood samples collected from both renal veins for cytokine-level measurements. EVs isolated from urine samples were characterized by imaging flow cytometry based on specific markers, including p16 (senescence marker), calyxin (podocytes), urate transporter 1 (proximal tubules), uromodulin (ascending limb of Henle's loop), and prominin-2 (distal tubules). Overall percentage of urinary p16+ EVs was elevated in EH and renovascular hypertension patients compared with healthy volunteers and correlated inversely with renal function and directly with renal vein cytokine levels. Urinary levels of p16+/urate transporter 1+ were elevated in all hypertensive subjects compared with healthy volunteers, whereas p16+/prominin-2+ levels were elevated only in EH versus healthy volunteers and p16+/uromodulin+ in renovascular hypertension versus EH. Conclusions Levels of p16+ EVs are elevated in urine of hypertensive patients and may reflect increased proximal tubular cellular senescence. In EH, EVs originate also from distal tubules and in renovascular hypertension from Henle's loop. Hence, urinary EVs levels may be useful to identify intrarenal sites of cellular senescence.
Collapse
Affiliation(s)
- Adrian Santelli
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
- Department of PhysiopathologyHospital de ClinicasMontevideoUruguay
| | - In O. Sun
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Alfonso Eirin
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | | | | | - Amir Lerman
- Department of Cardiovascular DiseasesMayo ClinicRochesterMN
| | | | | | | |
Collapse
|
16
|
Zheng J, Jiang Z, Chen D, Wang S, Zhao W, Li L. Pathological significance of urinary complement activation in diabetic nephropathy: A full view from the development of the disease. J Diabetes Investig 2019; 10:738-744. [PMID: 30239170 PMCID: PMC6497774 DOI: 10.1111/jdi.12934] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/30/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS/INTRODUCTION The aim of the present study was to obtain a full view of the changes of urinary complement activation products in the development of diabetic nephropathy and explore their possible significance in the disease process. MATERIALS AND METHODS A total of 62 patients at different stages of diabetic nephropathy, 20 diabetes patients without nephropathy and 20 healthy persons were enrolled. Urinary complement activation products, including C3a, C5a and C5b-9, were measured, and their associations with the progression of the disease were analyzed. RESULTS The urinary complement activation products increased markedly since the proteinuria stage, and were parallel with the progression of diabetic nephropathy. More severe renal tubular damage was observed in patients with higher levels of urinary complement activation products. The urinary complement activation products levels correlated closely with renal tubulointerstitial injury score and relative tubular interstitial volume. Multivariate regression analysis showed that elevated urinary complement activation products were independent risk factors for tubular injury in diabetic nephropathy patients. CONCLUSIONS Urinary complement activation might have a role in renal tubular interstitial injury in patients with diabetic nephropathy, especially in patients at a later stage of the disease.
Collapse
Affiliation(s)
- Jing‐Min Zheng
- Department of Nephrology Taizhou HospitalWenzhou Medical UniversityLihaiZhejiang ProvinceChina
- National Clinical Research Center of Kidney DiseasesJingling HospitalNanjing University School of MedicineNanjingChina
| | - Zuan‐Hong Jiang
- Department of Nephrology Taizhou HospitalWenzhou Medical UniversityLihaiZhejiang ProvinceChina
| | - De‐Jun Chen
- Department of Nephrology Taizhou HospitalWenzhou Medical UniversityLihaiZhejiang ProvinceChina
| | - Sa‐Sa Wang
- Department of Nephrology Taizhou HospitalWenzhou Medical UniversityLihaiZhejiang ProvinceChina
| | - Wen‐Jin Zhao
- National Clinical Research Center of Kidney DiseasesJingling HospitalNanjing University School of MedicineNanjingChina
| | - Li‐Juan Li
- National Clinical Research Center of Kidney DiseasesJingling HospitalNanjing University School of MedicineNanjingChina
| |
Collapse
|
17
|
|
18
|
Kelly CB, Hookham MB, Yu JY, Jenkins AJ, Nankervis AJ, Hanssen KF, Garg SK, Scardo JA, Hammad SM, Menard MK, Aston CE, Lyons TJ. Subclinical First Trimester Renal Abnormalities Are Associated With Preeclampsia in Normoalbuminuric Women With Type 1 Diabetes. Diabetes Care 2018; 41:120-127. [PMID: 29122892 PMCID: PMC5741157 DOI: 10.2337/dc17-1635] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/05/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study was conducted to determine the utility of tubular (urinary/plasma neutrophil gelatinase-associated lipocalin [NGAL] and urinary kidney injury molecule 1 [KIM-1]) and glomerular (estimated glomerular filtration rate [eGFR]) biomarkers in predicting preeclampsia (PE) in pregnant women with type 1 diabetes mellitus (T1DM) who were free of microalbuminuria and hypertension at the first trimester. RESEARCH DESIGN AND METHODS This was a prospective study of T1DM pregnancy. Maternal urinary and plasma NGAL, urinary KIM-1 (ELISA of frozen samples), and eGFR (Chronic Kidney Disease Epidemiology Collaboration equation) were determined at three study visits (V1: 12.4 ± 1.8; V2: 21.7 ± 1.4; V3: 31.4 ± 1.5 weeks' gestation [mean ± SD]) in 23 women with T1DM with subsequent PE (DM+PE+), 24 who remained normotensive (DM+PE-), and, for reference, in 19 normotensive pregnant women without diabetes (DM-). The groups with diabetes were matched for age, diabetes duration, and parity. All subjects were normotensive and free of microalbuminuria or albuminuria at V1. All study visits preceded the onset of PE. RESULTS Urinary creatinine-corrected NGAL (uNGALcc, ng/mg) was significantly elevated at V1 in DM+PE+ vs. DM+PE- women (P = 0.01); this remained significant after exclusion of leukocyte-positive samples (5 DM+PE+ and 2 DM+PE-) (P = 0.02). Accounting for BMI, HbA1c, and total daily insulin dose, a doubling of uNGALcc at V1 conferred a sevenfold increase in risk for PE (P = 0.026). In contrast, neither plasma NGAL nor urinary KIM-1 predicted PE. Also at V1, eGFR was elevated in DM+PE+ vs. DM+PE- (P = 0.04). CONCLUSIONS Early tubular and glomerular dysfunction may predict PE in first trimester women with T1DM, even if free of microalbuminuria. These data suggest that subclinical renal tubular and glomerular injury, if present early in pregnancy, may predispose women with T1DM to PE.
Collapse
Affiliation(s)
- Clare B Kelly
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, U.K.,Division of Endocrinology, Medical University of South Carolina, Charleston, SC
| | - Michelle B Hookham
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, U.K.,The Department of Clinical Biochemistry, Royal Victoria Hospital, Belfast, Northern Ireland, U.K
| | - Jeremy Y Yu
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, U.K.,Division of Endocrinology, Medical University of South Carolina, Charleston, SC
| | - Alicia J Jenkins
- Division of Endocrinology, Medical University of South Carolina, Charleston, SC.,National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Alison J Nankervis
- Diabetes Service, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Kristian F Hanssen
- Department of Endocrinology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Satish K Garg
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Denver, CO
| | | | - Samar M Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| | - M Kathryn Menard
- Division of Materno-Fetal Medicine, University of North Carolina, Chapel Hill, NC
| | - Christopher E Aston
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Timothy J Lyons
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, U.K. .,Division of Endocrinology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
19
|
Eltounali SA, Moodley J, Naicker T. Role of kidney biomarkers [Kidney injury molecule-1, Calbindin, Interleukin-18 and Monocyte chemoattractant protein-1] in HIV associated pre-eclampsia. Hypertens Pregnancy 2017; 36:288-294. [PMID: 29039974 DOI: 10.1080/10641955.2017.1385793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Both HIV infection and pre-eclampsia (PE) are associated with considerable maternal mortality in South Africa. This study was designed to compare the urinary levels of kidney injury molecule-1 (KIM-1), calbindin, interleukin-18 (IL-18), and monocyte chemoattractant protein-1 (MCP-1) in HIV associated normotensive and preeclamptic pregnancies. METHODS Following ethical approval and written consent, urine samples were collected from HIV negative (HIV -ve) normotensive pregnant (n = 19), HIV positive (HIV +ve) normotensive pregnant (n = 19), HIV -ve pre-eclamptic (n = 19) and HIV +ve pre-eclamptic (n = 19) women. The concentrations of KIM-1, calbindin, IL-18 and MCP-1 were assessed using the Bioplex technology. RESULTS In contrast to IL-18 (p > 0.05) and MCP-1 (p > 0.05), the concentrations of KIM-1 (p = 0.02) and calbindin (p = 0.02) were significantly higher in PE compared to normotensive pregnancies, irrespective of HIV status. Based on HIV status, all 4 analytes were similar between HIV+ve and HIV-ve groups. Urinary KIM-1 levels in the HIV -ve pre-eclamptics were significantly higher than those in the HIV -ve women with normal pregnancies (p = 0.007). The maternal hypertension and/or HIV profile has no marked impact on the fetal weight. CONCLUSION Our results demonstrate an increase in the urinary level of kidney injury molecule-1 and calbindin in PE, implicating their possible value as biomarkers of kidney injury. We observed no differences in the levels of KIM-1, IL-18, MCP-1 and calbindin based on HIV status. We propose that studies with larger sample sizes using these markers be conducted to establish their use as markers of diagnosing kidney injury in PE.
Collapse
Affiliation(s)
- Soumaya Abdullatif Eltounali
- a Optics and Imaging Centre, Nelson R. Mandela School of Medicine, College of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Jagidesa Moodley
- b Women's Health and HIV Research Unit, Nelson R. Mandela School of Medicine, College of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| | - Thajasvarie Naicker
- a Optics and Imaging Centre, Nelson R. Mandela School of Medicine, College of Health Sciences , University of KwaZulu-Natal , Durban , South Africa
| |
Collapse
|
20
|
Codsi E, Garovic VD, Gonzalez-Suarez ML, Milic N, Borowski KS, Rose CH, Davies NP, Kashani KB, Lieske JC, White WM. Longitudinal characterization of renal proximal tubular markers in normotensive and preeclamptic pregnancies. Am J Physiol Regul Integr Comp Physiol 2017; 312:R773-R778. [DOI: 10.1152/ajpregu.00509.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/29/2017] [Accepted: 04/18/2017] [Indexed: 01/10/2023]
Abstract
Glomerular damage is common in preeclampsia (PE), but the extent and etiology of tubular injury are not well understood. The aim of this study was to evaluate tubular injury in patients with PE and to assess whether it predates clinical disease. We performed a prospective cohort study of 315 pregnant women who provided urine samples at the end of the second trimester and at delivery. This analysis included women who developed PE ( n = 15), gestational hypertension (GH; n = 14), and normotensive controls (NC; n = 44). Urinary markers of tubular injury, α1-microglobulin (A1M), retinol-binding protein (RBP), kidney-injury molecule-1 (KIM1), complement C5b-9, tissue inhibitor metalloproteinase-2 (TIMP-2), and insulin-like growth factor binding protein-7 (IGFBP-7) were measured by enzyme-linked immunosorbent assay (ELISA) and reported in relation to urine creatinine concentration. Second-trimester concentrations of all markers were similar among groups. At delivery, A1M concentrations were higher in the PE group than in the GH and NC groups as an A1M/creatinine ratio >13 (66.7, 8.3, and 35%, respectively, P = 0.01). Concentrations of C5b-9 were higher in the PE group than in the GH and NC groups (medians 9.85, 0.05, and 0.28 ng/mg, respectively, P = 0.003). KIM1, RBP, TIMP-2, and IGFBP-7 concentrations did not differ among groups at delivery. In conclusion, proximal tubular dysfunction, as assessed by A1M and C5b-9, developed during the interval between the end of the second trimester and delivery in patients with PE. However, this was not matched by abnormalities in markers previously associated with tubular cell injury (KIM-1, IGFBP-7, and TIMP-2).
Collapse
Affiliation(s)
- Elisabeth Codsi
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vesna D. Garovic
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Maria L. Gonzalez-Suarez
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Natasa Milic
- Department of Medical Statistics and Informatics, University of Belgrade, Belgrade, Serbia
| | - Kristi S. Borowski
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Carl H. Rose
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Norman P. Davies
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kianoush B. Kashani
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - John C. Lieske
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Wendy M. White
- Department of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
21
|
Regal JF, Strehlke ME, Peterson JM, Wing CR, Parker JE, Nieto NF, Bemis LT, Gilbert JS, Fleming SD. Role of IgM and angiotensin II Type I receptor autoantibodies in local complement activation in placental ischemia-induced hypertension in the rat. Mol Immunol 2016; 78:38-47. [PMID: 27588825 DOI: 10.1016/j.molimm.2016.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 01/27/2023]
Abstract
Preeclampsia is characterized by development of hypertension during pregnancy and reduced placental perfusion. Previous studies in a rat model of placental ischemia-induced hypertension demonstrated that inhibiting complement activation attenuated increased maternal blood pressure with C3a and C5a identified as the important products of complement activation. Given that in other forms of ischemia both natural IgM and antigen antibody complexes initiate complement activation, we hypothesized that placental ischemia exposes neoepitopes recognized by IgM to cause local complement activation and hypertension. Alternatively, we postulated that autoantibody to angiotensin II Type 1 receptor (AT1-AA) interacts with AT1 receptors to cause complement activation. Since complement activation occurs in kidney and placenta in preeclampsia, we used immunohistochemistry to determine IgM deposition and local complement activation in each organ (C3 deposition), and quantitative real-time polymerase chain reaction (qRT-PCR) to quantitate mRNA for endogenous regulators of complement activation CD55, CD59 and Complement receptor 1-related gene/protein y (Crry). On gestation day (GD)14.5, timed pregnant Sprague Dawley rats underwent Sham surgery or placement of clips on inferior abdominal aorta and ovarian arteries to create placental ischemia using the reduced utero-placental perfusion pressure (RUPP) model. As previously reported, RUPP surgery increased mean arterial pressure and circulating C3a on GD19.5. In placenta, IgM and C3 deposition increased, whereas mRNA for complement regulators Crry and CD59 decreased along with Crry protein in RUPP compared to Sham treated animals. In kidney, IgM deposition increased in animals subjected to RUPP vs Sham surgery without a significant change in C3 deposition and coincident with an increase in mRNA for CD55 and CD59. The AT1 receptor antagonist losartan prevents placental ischemia-induced hypertension as well as AT1-AA interaction with AT1 receptors. However, losartan did not attenuate complement activation as measured by circulating C3a or placental C3 deposition. Importantly, our studies indicate that following placental ischemia, complement activation is not due to AT1-AA but is associated with IgM deposition. These studies suggest a role for natural antibodies interacting with placental ischemia-induced neoepitopes to activate complement and contribute to hypertension.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Megan E Strehlke
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Jenna M Peterson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Cameron R Wing
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Jordan E Parker
- Division of Biology, Kansas State University, Manhattan, KS, United States.
| | | | - Lynne T Bemis
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Jeffrey S Gilbert
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth campus, Duluth, MN, United States.
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
22
|
Mourad M, Jain J, Mehta MP, Feinberg BB, Burwick RM. Are We Getting Closer to Explaining Preeclampsia? CURRENT OBSTETRICS AND GYNECOLOGY REPORTS 2016. [DOI: 10.1007/s13669-016-0169-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Abstract
Our understanding of hypertension during pregnancy and, in particular, preeclampsia has changed dramatically over the last decade. During the last year (2014–2015), several articles published in Hypertension have provided important insights into the pathogenesis of preeclampsia and its related complications.1–38 In addition, Hypertension also published some key research communications that translated important basic science observations into the clinic. Some of these articles are briefly discussed, highlighting their significance to our understanding of the mechanism of the disease, to predict the disease and to treat or prevent hypertension during pregnancy and other preeclampsia-related complications.
Collapse
Affiliation(s)
- S Ananth Karumanchi
- From the Beth Israel Deaconess Medical Center, Boston, MA (S.A.K.); and Department of Physiology and Biophysics and the Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS (J.P.G.)
| | - Joey P Granger
- From the Beth Israel Deaconess Medical Center, Boston, MA (S.A.K.); and Department of Physiology and Biophysics and the Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS (J.P.G.).
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Preeclampsia is a gestational kidney disease characterized by glomerular endothelial injury, leading to maternal hypertension and proteinuria. If not addressed promptly, there is significant maternal and fetal morbidity and mortality. When severe, this disorder can cause hepatic and neurologic dysfunction. Understandably, this placental disease enters the focus of the obstetrician first; however, with progression, the nephrologist can also be enlisted. Typical complications include acute kidney injury, refractory hypertension, and acute pulmonary edema. This review summarizes recent literature on the pathogenesis of this condition and will highlight new diagnostic and therapeutic options for preeclampsia. RECENT FINDINGS Over the past decade, the role of soluble vascular factors in preeclampsia has shed light on the mechanism underlying this disease. During the last 2 years, several new therapeutics have been developed that target implicated circulating angiogenic factors, including soluble fms-like tyrosine kinase 1, an endogenous vascular endothelial growth factor inhibitor. Serum levels of angiogenic factors have been correlated with a constellation of hemodynamic and pathophysiologic changes. Thus, circulating levels of these factors may serve both diagnostic and prognostic purposes. SUMMARY Overall, our understanding of preeclampsia has developed significantly and the future holds promise for mechanism-based novel diagnostics and therapeutics.
Collapse
|
25
|
From Glomerular Endothelium to Podocyte Pathobiology in Preeclampsia: a Paradigm Shift. Curr Hypertens Rep 2015; 17:54. [DOI: 10.1007/s11906-015-0566-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
26
|
Regal JF, Gilbert JS, Burwick RM. The complement system and adverse pregnancy outcomes. Mol Immunol 2015; 67:56-70. [PMID: 25802092 DOI: 10.1016/j.molimm.2015.02.030] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/20/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023]
Abstract
Adverse pregnancy outcomes significantly contribute to morbidity and mortality for mother and child, with lifelong health consequences for both. The innate and adaptive immune system must be regulated to insure survival of the fetal allograft, and the complement system is no exception. An intact complement system optimizes placental development and function and is essential to maintain host defense and fetal survival. Complement regulation is apparent at the placental interface from early pregnancy with some degree of complement activation occurring normally throughout gestation. However, a number of pregnancy complications including early pregnancy loss, fetal growth restriction, hypertensive disorders of pregnancy and preterm birth are associated with excessive or misdirected complement activation, and are more frequent in women with inherited or acquired complement system disorders or complement gene mutations. Clinical studies employing complement biomarkers in plasma and urine implicate dysregulated complement activation in components of each of the adverse pregnancy outcomes. In addition, mechanistic studies in rat and mouse models of adverse pregnancy outcomes address the complement pathways or activation products of importance and allow critical analysis of the pathophysiology. Targeted complement therapeutics are already in use to control adverse pregnancy outcomes in select situations. A clearer understanding of the role of the complement system in both normal pregnancy and complicated or failed pregnancy will allow a rational approach to future therapeutic strategies for manipulating complement with the goal of mitigating adverse pregnancy outcomes, preserving host defense, and improving long term outcomes for both mother and child.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Jeffrey S Gilbert
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Richard M Burwick
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Oregon Health & Science University, Mail Code: L-458, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
27
|
Guseh SH, Feinberg BB, Dawood HY, Yamamoto HS, Fichorova RN, Burwick RM. Urinary Excretion of C5b-9 is Associated With the Anti-Angiogenic State in Severe Preeclampsia. Am J Reprod Immunol 2014; 73:437-44. [DOI: 10.1111/aji.12349] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/18/2014] [Indexed: 11/26/2022] Open
Affiliation(s)
- Stephanie H. Guseh
- Department of Obstetrics, Gynecology and Reproductive Biology; Brigham and Women's Hospital; Harvard Medical School; Boston MA USA
| | - Bruce B. Feinberg
- Division of Maternal Fetal Medicine; Department of Obstetrics and Gynecology; Columbia University Medical Center; New York NY USA
| | - Hassan Y. Dawood
- Department of Obstetrics, Gynecology and Reproductive Biology; Laboratory of Genital Tract Biology; Brigham and Women's Hospital; Harvard Medical School; Boston MA USA
| | - Hidemi S. Yamamoto
- Department of Obstetrics, Gynecology and Reproductive Biology; Laboratory of Genital Tract Biology; Brigham and Women's Hospital; Harvard Medical School; Boston MA USA
| | - Raina N. Fichorova
- Department of Obstetrics, Gynecology and Reproductive Biology; Laboratory of Genital Tract Biology; Brigham and Women's Hospital; Harvard Medical School; Boston MA USA
| | - Richard M. Burwick
- Division of Maternal Fetal Medicine; Department of Obstetrics and Gynecology; Oregon Health and Science University; Portland OR USA
| |
Collapse
|