1
|
Aslanian-Kalkhoran L, Nouri N, Soltani-Zangbar MS, Mardi A, Aghebati-Maleki L. Immunoglobulin therapy for infertility and the role of immune cells in pregnancy success: An extensive investigation and update. J Reprod Immunol 2025; 169:104458. [PMID: 40015106 DOI: 10.1016/j.jri.2025.104458] [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: 12/31/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
In the United States, roughly one out of every eight couples, or 7.5 million women, experience challenges related to conceiving or maintaining a pregnancy. The body's immune response is vital during pregnancy. T cells, natural killer (NK) cells, B cells, and macrophages (MQ) are immune cells in the female reproductive tract. They are in charge of maintaining tissue homeostasis and regulating the immune system's response to invasive pathogens. Failure to regulate these immune cells might result in inflammation, which reduces fertility. The immune system modulation of pregnancy loss has been studied with intralipid, intravenous immunoglobulin (IVIG), and paternal leukocyte vaccination. A concentrated antibody called intravenous immunoglobulin (IVIG) is utilized as a biological agent to treat autoimmune, viral, and inflammatory diseases and some immunodeficiencies. The main objective of this treatment is to restore a damaged immune system. IgGs, through binding to specific antigens, promote the innate immunity's cellular and humoral immune response by activating complements and binding to Fc receptors of several immune cells. Contrariwise, IVIG regulates pathogenic autoimmunity in animal models, including skin-blister diseases, nephrotoxic nephritis, and K/BxN arthritis. IVIG has, therefore, been of great interest as an immune modulator in several immune disorders. This review aims to investigate the immunological reasons of reproductive failure, focusing on the immunomodulatory effects of IVIG in its treatment.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narjes Nouri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Mittal R, Prasad K, Lemos JRN, Arevalo G, Hirani K. Unveiling Gestational Diabetes: An Overview of Pathophysiology and Management. Int J Mol Sci 2025; 26:2320. [PMID: 40076938 PMCID: PMC11900321 DOI: 10.3390/ijms26052320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is characterized by an inadequate pancreatic β-cell response to pregnancy-induced insulin resistance, resulting in hyperglycemia. The pathophysiology involves reduced incretin hormone secretion and signaling, specifically decreased glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), impairing insulinotropic effects. Pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), impair insulin receptor substrate-1 (IRS-1) phosphorylation, disrupting insulin-mediated glucose uptake. β-cell dysfunction in GDM is associated with decreased pancreatic duodenal homeobox 1 (PDX1) expression, increased endoplasmic reticulum stress markers (CHOP, GRP78), and mitochondrial dysfunction leading to impaired ATP production and reduced glucose-stimulated insulin secretion. Excessive gestational weight gain exacerbates insulin resistance through hyperleptinemia, which downregulates insulin receptor expression via JAK/STAT signaling. Additionally, hypoadiponectinemia decreases AMP-activated protein kinase (AMPK) activation in skeletal muscle, impairing GLUT4 translocation. Placental hormones such as human placental lactogen (hPL) induce lipolysis, increasing circulating free fatty acids which activate protein kinase C, inhibiting insulin signaling. Placental 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) overactivity elevates cortisol levels, which activate glucocorticoid receptors to further reduce insulin sensitivity. GDM diagnostic thresholds (≥92 mg/dL fasting, ≥153 mg/dL post-load) are lower than type 2 diabetes to prevent fetal hyperinsulinemia and macrosomia. Management strategies focus on lifestyle modifications, including dietary carbohydrate restriction and exercise. Pharmacological interventions, such as insulin or metformin, aim to restore AMPK signaling and reduce hepatic glucose output. Emerging therapies, such as glucagon-like peptide-1 receptor (GLP-1R) agonists, show potential in improving glycemic control and reducing inflammation. A mechanistic understanding of GDM pathophysiology is essential for developing targeted therapeutic strategies to prevent both adverse pregnancy outcomes and the progression to overt diabetes in affected women.
Collapse
Affiliation(s)
| | | | | | | | - Khemraj Hirani
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (K.P.); (J.R.N.L.); (G.A.)
| |
Collapse
|
3
|
Yu W, Miao H, Gong Y. Lymphocyte Subsets and Cytokine Changes in Women With Gestational Diabetes Mellitus: A Systematic Review. J Diabetes Res 2025; 2025:3494697. [PMID: 40225013 PMCID: PMC11986944 DOI: 10.1155/jdr/3494697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/28/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: Gestational diabetes mellitus (GDM) is a major health concern during pregnancy, affecting both the mother and the baby. Immune system alterations, particularly changes in lymphocyte subsets and cytokine profiles, have been associated with the pathophysiology of various metabolic disorders, including diabetes. This study is aimed at systematically reviewing the literature on the changes in lymphocyte subsets and cytokines in GDM. Methods: In this systematic review, we applied specific criteria to select observational studies (such as case-controls, cross-sectionals, or cohorts) that focused on pregnant women. We performed an extensive search across electronic databases, including Web of Science, Scopus, PubMed, MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and Google Scholar, from January 1, 2010, to March 20, 2024. Results: A total of 19 articles, with 2517 participants (1128 with GDM and 1389 without GDM), were included in the qualitative synthesis. Due to high heterogeneity among the articles, a meta-analysis was not conducted. The studies assessed 35 different lymphocyte subsets or proportions. The most commonly assessed subsets were CD3+ T cell (five articles, mostly no difference between GDM and non-GDM), CD4+ T cell (five articles with contradictory results), CD8+ T cell (four articles with contradictory results), B cell and NK cell (three articles, mostly no difference between GDM and non-GDM), and Tregs (three articles with contradictory results). Additionally, 32 cytokines or proportions were assessed in the studies. The most commonly assessed cytokines were IL-6 (eight articles, higher or similar levels in GDM compared to non-GDM), TNF-α (seven articles, mostly higher or similar levels in GDM compared to non-GDM), IL-10 (six articles, mostly no difference between GDM and non-GDM), IL-2 (three articles, mostly no difference between GDM and non-GDM), and IFN-γ (three articles with contradictory results). Conclusion: According to the results, there were no significant changes in CD3+ T cells, B cells, NK cells, IL-10, and IL-2 in GDM. However, the levels of IL-6 and TNF-α were higher or similar in GDM compared to non-GDM. The changes of other lymphocyte subsets and cytokines in GDM remained unclear.
Collapse
Affiliation(s)
- Wang Yu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Huang Miao
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Yunhui Gong
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Sebina I, Bidgood C, Stalley F, Hartel G, Stark T, Callaway L, Amoako A, Lehner C, Dekker Nitert M, Phipps S. Pre-pregnancy obesity is associated with an altered maternal metabolome and reduced Flt3L expression in preterm birth. Sci Rep 2024; 14:30027. [PMID: 39627409 PMCID: PMC11615298 DOI: 10.1038/s41598-024-81194-4] [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: 03/14/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Mechanisms linking pre-pregnancy obesity to increased preterm birth risk are unclear. Here, we examined the impact of pre-pregnancy obesity on metabolites, Fms-related tyrosine kinase 3 ligand (Flt3L), and proinflammatory cytokine profiles in preterm birth. We used cytokine bead array, ELISA and Gas Chromatography-Mass Spectrometry (GC-MS) to determine cytokine and metabolite profiles in maternal and cord blood samples from 124 pregnant women in Australia, who gave birth at term (n = 86) or preterm (n = 38). Besides the expected variations in birth weight and gestational age, all demographic characteristics, including pre-pregnancy body mass index, were similar between the term and preterm birth groups. Mothers in the preterm birth group had reduced Flt3L (P = 0.002) and elevated IL-6 (P = 0.002) compared with term birthing mothers. Among mothers who gave birth preterm, those with pre-pregnancy obesity had lower Flt3L levels (P = 0.02) compared with lean mothers. Flt3L and IL-6 were similar in cord blood across both groups, but TNFα levels (P = 0.02) were reduced in preterm newborns. Metabolomic analysis revealed significant shifts in essential metabolites in women with pre-pregnancy obesity, some of which were linked to preterm births. Our findings suggest that maternal pre-pregnancy obesity alters the metabolome and reduces Flt3L expression, potentially increasing risk of preterm birth.
Collapse
Affiliation(s)
- Ismail Sebina
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia.
| | - Charles Bidgood
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia
| | - Felicity Stalley
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Gunter Hartel
- Statistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Public Health, The University of Queensland, Brisbane, QLD, Australia
| | - Terra Stark
- Metabolomics Australia (Queensland Node), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Leonie Callaway
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Akwasi Amoako
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Christoph Lehner
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon Phipps
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia
| |
Collapse
|
5
|
Yang H. Gut Microbiota, Circulating Metabolites and Risk of Endometriosis: A Two-Step Mendelian Randomization Study. Pol J Microbiol 2024; 73:491-503. [PMID: 39670637 PMCID: PMC11639408 DOI: 10.33073/pjm-2024-041] [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: 06/30/2024] [Accepted: 09/19/2024] [Indexed: 12/14/2024] Open
Abstract
Epidemiological studies and animal models have suggested a possible link between gut microbiota (GM), circulating metabolites, and endometriosis (EMs) pathogenesis. However, whether these associations are causal or merely due to confounding factors remains unclear. We conducted a two-sample and two-step Mendelian randomization (MR) study to elucidate the potential causal relationship between GM and EMs, and the mediating role of circulating metabolites. Our MR analysis revealed that higher abundances of class Negativicutes, and order Selenomonadales, as well as genera Dialister, Enterorhabdus, Eubacterium xylanophilum group, Methanobrevibacter were associated with an increased risk of EMs (Odds Ratio (OR) range: 1.0019-1.0037). Conversely, higher abundances of genera Coprococcus 1 and Senegalimassilia were linked to reduced risk of EMs (OR range: 0.9964-0.9967). Additionally, elevated levels of circulating metabolites such as 1-eicosatrienoyl-glycerophosphocholine and 1-oleoylglycerophosphocholine were found to be associated with heightened risk of EMs (OR range: 2.21-3.16), while higher concentrations of 3-phenylpropionate and dihomo-linolenate were protective (OR range: 0.285-0.535). Two-step MR analysis indicated that specific microbial taxa, notably genus Enterorhabdus and order Selenomonadales, might function as mediators linking circulating metabolites to the risk of EMs. Our findings suggest a probable causal relationship between GM, circulating metabolites, and EMs, indicating that GM may mediate the influence of circulating metabolites on the pathophysiology of EMs. These results offer new leads for future mechanistic studies and could inform clinical translational research.
Collapse
Affiliation(s)
- Hua Yang
- Department of Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
6
|
Yang H. The causality between gut microbiota and endometriosis: a bidirectional Mendelian randomization study. Front Med (Lausanne) 2024; 11:1434582. [PMID: 39650192 PMCID: PMC11621931 DOI: 10.3389/fmed.2024.1434582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
Background Observational studies and animal experiments had suggested a potential relationship between gut microbiota abundance and pathogenesis of endometriosis (EMs), but the relevance of this relationship remains to be clarified. Methods We perform a two-sample bidirectional Mendelian randomization (MR) analysis to explore whether there is a causal correlation between the abundance of the gut microbiota and EMs and the direction of causality. Genome-wide association study (GWAS) data ukb-d-N80, finn-b-N14-EM, and MiBinGen were selected. Inverse variance weighted (IVW), weighted median, and MR Egger are selected for causal inference. The Cochran Q test, Egger intercept test, and leave-one-out analysis are performed for sensitivity analyses. Results In the primary outcome, we find that a higher abundance of class Negativicutes, genus Dialister, genus Enterorhabdus, genus Eubacterium xylanophilum group, genus Methanobrevibacter and order Selenomonadales predict a higher risk of EMs, and a higher abundance of genus Coprococcus and genus Senegalimassilia predict a lower risk of EMs. During verifiable outcomes, we find that a higher abundance of phylum Cyanobacteria, genus Ruminococcaceae UCG002, and genus Coprococcus 3 predict a higher risk of EMs, and a higher abundance of genus Flavonifracto, genus Bifidobacterium, and genus Rikenellaceae RC9 predict a lower risk of EMs. In primary reverse MR analysis, we find that EMs predict a lower abundance of the genus Eubacterium fissicatena group, genus Prevotella7, genus Butyricicoccus, family Lactobacillaceae, and a higher abundance of genus Ruminococcaceae UCG009. In verifiable reverse MR analysis, we find that EMs predict a lower abundance of the genus Ruminococcaceae UCG004 and a higher abundance of the genus Howardella. Conclusion Our study implies a mutual causality between gut microbiota abundance and the pathogenesis of EMs, which may provide a novel direction for EMs diagnosis, prevention, and treatment, may promote future functional or clinical analysis.
Collapse
Affiliation(s)
- Hua Yang
- Department of Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
7
|
Wu Z, Hou Q, Chi H, Liu J, Mei Y, Chen T, Yang K, Zheng J, Xu J, Wei F, Wang L. Single-cell RNA sequencing reveals a distinct profile of bone immune microenvironment and decreased osteoclast differentiation in type 2 diabetic mice. Genes Dis 2024; 11:101145. [PMID: 39281831 PMCID: PMC11399629 DOI: 10.1016/j.gendis.2023.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/07/2023] [Accepted: 09/16/2023] [Indexed: 09/18/2024] Open
Abstract
The pathogenic effects of type 2 diabetes on bone tissue are gaining attention, but the cellular and molecular mechanisms underlying osteoimmunology are still unclear in diabetes-related bone diseases. We delineated the single-cell transcriptome of bone marrow cells from both wide type and type 2 diabetes mice, which provided the first detailed global profile of bone marrow cells and revealed a distinct bone immune microenvironment at the genetic level under type 2 diabetic condition. It was observed that osteoclast activity was inhibited due to a dysregulated cytokine network, which ultimately led to decreased osteoclast formation and differentiation. In type 2 diabetes mice, a specific C d 36 + cluster (cluster 18, monocytes/macrophages 2) was identified as the precursor of osteoclasts with diminished differentiation potential. AP-1 was demonstrated to be the key transcription factor in the underlying mechanism.
Collapse
Affiliation(s)
- Zimei Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Department of Orthopedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Qiaodan Hou
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Heng Chi
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jihong Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| | - Yixin Mei
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tingting Chen
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Kunkun Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jingna Zheng
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jing Xu
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| | - Fuxin Wei
- Department of Orthopedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Lin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Southern University of Science and Technology Hospital, Shenzhen, Guangdong 518055, China
| |
Collapse
|
8
|
Razo-Azamar M, Nambo-Venegas R, Quevedo IR, Juárez-Luna G, Salomon C, Guevara-Cruz M, Palacios-González B. Early-Pregnancy Serum Maternal and Placenta-Derived Exosomes miRNAs Vary Based on Pancreatic β-Cell Function in GDM. J Clin Endocrinol Metab 2024; 109:1526-1539. [PMID: 38127956 DOI: 10.1210/clinem/dgad751] [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: 10/06/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023]
Abstract
CONTEXT Pancreatic β-cell function impairment is a key mechanism for developing gestational diabetes mellitus (GDM). Maternal and placental exosomes regulate maternal and placental responses during hyperglycemia. Studies have associated exosomal micro-RNAs (miRNAs) with GDM development. To date, no studies have been reported that evaluate the profile of miRNAs present in maternal and placental exosomes in the early stages of gestation from pregnancies that develop GDM. OBJECTIVE We assessed whether early-pregnancy serum maternal and placenta-derived exosomes miRNA profiles vary according to pancreatic β-cell function in women who will develop GDM. METHODS A prospective nested case-control study was used to identify exosomal miRNAs that vary in early-pregnancy stages (<18 weeks of gestation) from women with normoglycemia and those who developed GDM based on their pancreatic β-cell function using the homeostasis model assessment of pancreatic β-cell function (HOMA-%β) index. Early-pregnancy serum maternal and placenta-derived exosomes were isolated to obtain miRNA profiles. Potential target and pathway analyses were performed to identify molecular and metabolic pathways associated with the exosomal miRNAs identified. RESULTS In early-pregnancy stages, serum maternal exosome size and concentration are modified in GDM group and fluctuate according to HOMA-%β index. Serum maternal exosomal hsa-miR-149-3p and hsa-miR-455-3p in GDM are related to insulin secretion and signaling, lipolysis, and adipocytokine signaling. Early-pregnancy serum placenta-derived exosomes hsa-miR-3665 and hsa-miR-6727-5p in GDM are related to regulating genes involved in response to immunological tolerance of pregnancy and pathways associated with placental dysfunction. CONCLUSION Early serum exosomal miRNAs differ depending on their origin (maternal or placental) and pancreatic β-cell function. This research provides insights into the interactions between maternal and placental exosomal miRNAs and may have implications for identifying potential biomarkers or therapeutic targets for GDM.
Collapse
Affiliation(s)
- Melissa Razo-Azamar
- Laboratorio de Envejecimiento Saludable del Instituto Nacional de Medicina Genómica (INMEGEN) en el Centro de Investigación sobre Envejecimiento (CIE-CINVESTAV Sede Sur), 14330 CDMX, México
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080 CDMX, México
| | - Rafael Nambo-Venegas
- Laboratorio de Bioquímica de Enfermedades Crónicas Instituto Nacional de Medicina Genómica (INMEGEN), 14610 CDMX, México
| | - Iván Rafael Quevedo
- Departamento de Ingeniería Química Industrial y de Alimentos (DIQIA), Universidad Iberoamericana Ciudad de México (UIA), 01219 CDMX, México
| | - Gregorio Juárez-Luna
- Departamento de Ingeniería Química Industrial y de Alimentos (DIQIA), Universidad Iberoamericana Ciudad de México (UIA), 01219 CDMX, México
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, 8320000 Santiago, Chile
| | - Martha Guevara-Cruz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080 CDMX, México
| | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable del Instituto Nacional de Medicina Genómica (INMEGEN) en el Centro de Investigación sobre Envejecimiento (CIE-CINVESTAV Sede Sur), 14330 CDMX, México
| |
Collapse
|
9
|
Ji Z, Zhang C, Yuan J, He Q, Zhang X, Yang D, Xu N, Chu J. Is there a causal association between gestational diabetes mellitus and immune mediators? A bidirectional Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1358144. [PMID: 38706698 PMCID: PMC11066251 DOI: 10.3389/fendo.2024.1358144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/19/2024] [Indexed: 05/07/2024] Open
Abstract
Background Diabetes that only appears or is diagnosed during pregnancy is referred to as gestational diabetes mellitus (GDM). The maternal physiological immune profile is essential for a positive pregnancy outcome. However, the causal relationship between GDM and immunophenotypes is not fully defined. Methods Based on the high-density genetic variation data at the genome-wide level, we evaluated the logical associations between 731 specific immune mediators and GDM using bidirectional Mendelian randomization (MR). The inverse variance weighted (IVW) was the main method employed for MR analysis. We performed multiple methods to verify the robustness and dependability of the MR results, and sensitivity measures were applied to rule out potential heterogeneity and horizontal pleiotropy. Results A substantial causal association between several immune mediators and GDM was detected. After FDR testing, HLA DR++ monocyte %leukocyte and HLA DR on plasmacytoid DC were shown to increase the risk of GDM; in contrast, CD127 on CD28+ CD45RA+ CD8br and CD19 on PB/PC were shown to attenuate the effect of GDM. Moreover, the progression of GDM has been shown to decrease the maternal levels of CD39+ activated Treg AC, CD39+ activated Treg %CD4 Treg, CD39+ resting Treg AC, CD39+ resting Treg %CD4 Treg, and CD39+ CD8BR %T cell. Conclusions Our findings support a possible causal association between GDM and various immunophenotypes, thus facilitating the provision of multiple options for preventive recognition as well as for the diagnostic and therapeutic management of GDM in clinical practice.
Collapse
Affiliation(s)
- Zhangxin Ji
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chenxu Zhang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jingjing Yuan
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Research and Technology Center, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Qing He
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xinyu Zhang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Dongmei Yang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Na Xu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and International Joint Laboratory on Tea Chemistry and Health Effects of Ministry of Education, Anhui Agricultural University, Hefei, Anhui, China
| | - Jun Chu
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Research and Technology Center, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Surgery, Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
10
|
Xiong Y, Wang Y, Wu M, Chen S, Lei H, Mu H, Yu H, Hou Y, Tang K, Chen X, Dong J, Wang X, Chen L. Aberrant NK cell profile in gestational diabetes mellitus with fetal growth restriction. Front Immunol 2024; 15:1346231. [PMID: 38375483 PMCID: PMC10875967 DOI: 10.3389/fimmu.2024.1346231] [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: 11/29/2023] [Accepted: 01/17/2024] [Indexed: 02/21/2024] Open
Abstract
Gestational diabetes mellitus (GDM) is a gestational disorder characterized by hyperglycemia, that can lead to dysfunction of diverse cells in the body, especially the immune cells. It has been reported that immune cells, specifically natural killer (NK) cells, play a crucial role in normal pregnancy. However, it remains unknown how hyperglycemia affects NK cell dysfunction thus participates in the development of GDM. In this experiment, GDM mice were induced by an intraperitoneal injection of streptozotocin (STZ) after pregnancy and it has been found that the intrauterine growth restriction occurred in mice with STZ-induced GDM, accompanied by the changed proportion and function of NK cells. The percentage of cytotoxic CD27-CD11b+ NK cells was significantly increased, while the proportion of nourished CD27-CD11b- NK cells was significantly reduced in the decidua of GDM mice. Likewise, the same trend appeared in the peripheral blood NK cell subsets of GDM patients. What's more, after intrauterine reinfusion of NK cells to GDM mice, the fetal growth restriction was alleviated and the proportion of NK cells was restored. Our findings provide a theoretical and experimental basis for further exploring the pathogenesis of GDM.
Collapse
Affiliation(s)
- Yujing Xiong
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yazhen Wang
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
| | - Mengqi Wu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shuqiang Chen
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Hui Lei
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Hui Mu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Haikun Yu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Yongli Hou
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
| | - Kang Tang
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xutao Chen
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
| | - Jie Dong
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Xiaohong Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Lihua Chen
- Department of Immunology, Air Force Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
11
|
Mercado-Evans V, Mejia ME, Zulk JJ, Ottinger S, Hameed ZA, Serchejian C, Marunde MG, Robertson CM, Ballard MB, Ruano SH, Korotkova N, Flores AR, Pennington KA, Patras KA. Gestational diabetes augments group B Streptococcus infection by disrupting maternal immunity and the vaginal microbiota. Nat Commun 2024; 15:1035. [PMID: 38310089 PMCID: PMC10838280 DOI: 10.1038/s41467-024-45336-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/19/2024] [Indexed: 02/05/2024] Open
Abstract
Group B Streptococcus (GBS) is a pervasive perinatal pathogen, yet factors driving GBS dissemination in utero are poorly defined. Gestational diabetes mellitus (GDM), a complication marked by dysregulated immunity and maternal microbial dysbiosis, increases risk for GBS perinatal disease. Using a murine GDM model of GBS colonization and perinatal transmission, we find that GDM mice display greater GBS in utero dissemination and subsequently worse neonatal outcomes. Dual-RNA sequencing reveals differential GBS adaptation to the GDM reproductive tract, including a putative glycosyltransferase (yfhO), and altered host responses. GDM immune disruptions include reduced uterine natural killer cell activation, impaired recruitment to placentae, and altered maternofetal cytokines. Lastly, we observe distinct vaginal microbial taxa associated with GDM status and GBS invasive disease status. Here, we show a model of GBS dissemination in GDM hosts that recapitulates several clinical aspects and identifies multiple host and bacterial drivers of GBS perinatal disease.
Collapse
Affiliation(s)
- Vicki Mercado-Evans
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marlyd E Mejia
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jacob J Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Samantha Ottinger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zainab A Hameed
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Camille Serchejian
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Madelynn G Marunde
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Clare M Robertson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mallory B Ballard
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Simone H Ruano
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Natalia Korotkova
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Anthony R Flores
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School, UTHealth Houston, Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Kathleen A Pennington
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Arain H, Patel T, Mureanu N, Efthymiou A, Lombardi G, Tree T, Nicolaides KH, Shangaris P. Regulatory T cells in the peripheral blood of women with gestational diabetes: a systematic review and meta-analysis. Front Immunol 2023; 14:1226617. [PMID: 38111588 PMCID: PMC10726109 DOI: 10.3389/fimmu.2023.1226617] [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: 05/21/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Background Gestational diabetes (GDM) affects approximately 14% of pregnancies globally and is associated with short- and long-term complications for both the mother and child. In addition, GDM has been linked to chronic low-grade inflammation with recent research indicating a potential immune dysregulation in pathophysiology and a disparity in regulatory T cells. Objective This systematic review and meta-analysis aimed to determine whether there is an association between GDM and the level of Tregs in the peripheral blood. Methods Literature searches were conducted in PubMed, Embase, and Ovid between the 7th and 14th of February 2022. The inclusion criteria were any original studies published in the English language, measuring differentiated Tregs in women with GDM compared with glucose-tolerant pregnant women. Meta-analysis was performed between comparable Treg markers. Statistical tests were used to quantify heterogeneity: τ 2, χ 2, and I 2. Study quality was assessed using a modified version of the Newcastle-Ottawa scale. Results The search yielded 223 results: eight studies were included in the review and seven in the meta-analysis (GDM = 228, control = 286). Analysis of Tregs across all trimesters showed significantly lower Treg numbers in women with GDM (SMD, -0.76; 95% CI, -1.37, -0.15; I 2 = 90%). This was reflected in the analysis by specific Treg markers (SMD -0.55; 95% CI, -1.04, -0.07; I 2 = 83%; third trimester, five studies). Non-significant differences were found within subgroups (differentiated by CD4+FoxP3+, CD4+CD127-, and CD4+CD127-FoxP3) of both analyses. Conclusion GDM is associated with lower Treg numbers in the peripheral maternal blood. In early pregnancy, there is clinical potential to use Treg levels as a predictive tool for the subsequent development of GDM. There is also a potential therapeutic intervention to prevent the development of GDM by increasing Treg populations. However, the precise mechanism by which Tregs mediate GDM remains unclear. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42022309796.
Collapse
Affiliation(s)
- Hania Arain
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
| | - Tina Patel
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
| | - Nicoleta Mureanu
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, United Kingdom
| | - Athina Efthymiou
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, United Kingdom
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Timothy Tree
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Kypros H. Nicolaides
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, United Kingdom
| | - Panicos Shangaris
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine King’s College London, London, United Kingdom
- Harris Birthright Research Centre for Fetal Medicine, King’s College Hospital, London, United Kingdom
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
13
|
Singh P, Elhaj DAI, Ibrahim I, Abdullahi H, Al Khodor S. Maternal microbiota and gestational diabetes: impact on infant health. J Transl Med 2023; 21:364. [PMID: 37280680 PMCID: PMC10246335 DOI: 10.1186/s12967-023-04230-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a common complication of pregnancy that has been associated with an increased risk of obesity and diabetes in the offspring. Pregnancy is accompanied by tightly regulated changes in the endocrine, metabolic, immune, and microbial systems, and deviations from these changes can alter the mother's metabolism resulting in adverse pregnancy outcomes and a negative impact on the health of her infant. Maternal microbiomes are significant drivers of mother and child health outcomes, and many microbial metabolites are likely to influence the host health. This review discusses the current understanding of how the microbiota and microbial metabolites may contribute to the development of GDM and how GDM-associated changes in the maternal microbiome can affect infant's health. We also describe microbiota-based interventions that aim to improve metabolic health and outline future directions for precision medicine research in this emerging field.
Collapse
Affiliation(s)
- Parul Singh
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- Research Department, Sidra Medicine, Doha, Qatar
| | | | - Ibrahim Ibrahim
- Women's Department, Sidra Medicine, Weill Cornell Medical College-Qatar, Doha, Qatar
| | - Hala Abdullahi
- Women's Department, Sidra Medicine, Weill Cornell Medical College-Qatar, Doha, Qatar
| | - Souhaila Al Khodor
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.
- Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
14
|
Hua S, Wang S, Cai J, Wu L, Cao Y. Myeloid-derived suppressor cells: Are they involved in gestational diabetes mellitus? Am J Reprod Immunol 2023:e13711. [PMID: 37157925 DOI: 10.1111/aji.13711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is currently the most common metabolic complication during pregnancy, with an increasing prevalence worldwide. Maternal immune dysregulation might be partly responsible for the pathophysiology of GDM. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells, emerging as a new immune regulator with potent immunosuppressive capacity. Although the fate and function of these cells were primarily described in pathological conditions such as cancer and infection, accumulating evidences have spotlighted their beneficial roles in homeostasis and physiological conditions. Recently, several studies have explored the roles of MDSCs in the diabetic microenvironment. However, the fate and function of these cells in GDM are still unknown. The current review summarized the existing knowledges about MDSCs and their potential roles in diabetes during pregnancy in an attempt to highlight our current understanding of GDM-related immune dysregulation and identify areas where further study is required.
Collapse
Affiliation(s)
- Siyu Hua
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Shanshan Wang
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinyang Cai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lamei Wu
- Department of Perinatal Healthcare, Huai'an District Maternity and Child Health Hospital, Huai'an, Jiangsu, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
França DCH, França EL, Sobrevia L, Barbosa AMP, Honorio-França AC, Rudge MVC. Integration of nutrigenomics, melatonin, serotonin and inflammatory cytokines in the pathophysiology of pregnancy-specific urinary incontinence in women with gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166737. [PMID: 37146917 DOI: 10.1016/j.bbadis.2023.166737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023]
Abstract
Gestational diabetes mellitus is an important public health problem and has been associated with the development of pregnancy-specific urinary incontinence. The interaction is related to hyperglycemia, and inflammatory and hormonal patterns, which favor functional alterations in different organs and systems. Several genes associated with human diseases have been identified and partially characterized. Most of these genes are known to cause monogenic diseases. However, about 3 % of diseases do not fit the monogenic theory due to the complex interactions between multiple genes and environmental factors, as in chronic metabolic diseases such as diabetes. The nutritional, immunological, and hormonal patterns associated with changes in maternal metabolism may influence and contribute to greater susceptibility to urinary tract disorders. However, early systematic reviews have not yielded consistent findings for these associations. This literature review summarizes important new findings from integrating nutrigenomics, hormones, and cytokines in women with Gestational diabetes mellitus and pregnancy-specific urinary incontinence. Changes in maternal metabolism due to hyperglycemia can generate an inflammatory environment with increased inflammatory cytokines. This environment modulated by inflammation can alter tryptophan uptake through food and thus influence the production of serotonin and melatonin. As these hormones seem to have protective effects against smooth muscle dysfunction and to restore the impaired contractility of the detrusor muscle, it is assumed that these changes may favor the onset of urinary incontinence specific to pregnancy.
Collapse
Affiliation(s)
- Danielle Cristina Honorio França
- Department of Gynecology and Obstetrics, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-687, Brazil.
| | - Eduardo Luzía França
- Institute of Biological and Health Science, Federal University of Mato Grosso (UFMT), Barra do Garças 78605-091, Brazil.
| | - Luis Sobrevia
- Department of Gynecology and Obstetrics, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, E-41012 Seville, Spain; Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Australia; Department of Pathology and Medical Biology, University of Groningen, 9713GZ Groningen, the Netherlands; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey 64710, Mexico.
| | - Angélica Mércia Pascon Barbosa
- Department of Gynecology and Obstetrics, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-687, Brazil; Department of Physiotherapy and Occupational Therapy, School of Philosophy and Sciences, São Paulo State University (UNESP), Marilia 17525-900, Brazil
| | | | - Marilza Vieira Cunha Rudge
- Department of Gynecology and Obstetrics, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu 18618-687, Brazil.
| |
Collapse
|
16
|
Zhu Y, Liu X, Xu Y, Lin Y. Hyperglycemia disturbs trophoblast functions and subsequently leads to failure of uterine spiral artery remodeling. Front Endocrinol (Lausanne) 2023; 14:1060253. [PMID: 37091848 PMCID: PMC10113679 DOI: 10.3389/fendo.2023.1060253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Uterine spiral artery remodeling is necessary for fetal growth and development as well as pregnancy outcomes. During remodeling, trophoblasts invade the arteries, replace the endothelium and disrupt the vascular smooth muscle, and are strictly regulated by the local microenvironment. Elevated glucose levels at the fetal-maternal interface are associated with disorganized placental villi and poor placental blood flow. Hyperglycemia disturbs trophoblast proliferation and invasion via inhibiting the epithelial-mesenchymal transition, altering the protein expression of related proteases (MMP9, MMP2, and uPA) and angiogenic factors (VEGF, PIGF). Besides, hyperglycemia influences the cellular crosstalk between immune cells, trophoblast, and vascular cells, leading to the failure of spiral artery remodeling. This review provides insight into molecular mechanisms and signaling pathways of hyperglycemia that influence trophoblast functions and uterine spiral artery remodeling.
Collapse
Affiliation(s)
- Yueyue Zhu
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yi Lin,
| |
Collapse
|
17
|
Oliveira-Scussel ACDM, Ferreira PTM, Resende RDS, Ratkevicius-Andrade CM, Gomes ADO, Paschoini MC, De Vito FB, Farnesi-de-Assunção TS, da Silva MV, Mineo JR, Rodrigues DBR, Rodrigues V. Association of gestational diabetes mellitus and negative modulation of the specific humoral and cellular immune response against Toxoplasma gondii. Front Immunol 2022; 13:925762. [PMID: 36203592 PMCID: PMC9531261 DOI: 10.3389/fimmu.2022.925762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
In order to evaluate and compare the specific immune response of pregnant women (PW) chronically infected with Toxoplasma gondii, with and without gestational diabetes mellitus (GDM), and the humoral response of their respective newborns (NB), the study was carried out on 81 PW (34 GDM and 47 controls) from whose medical records the results of the oral glucose tolerance test (OGTT) were obtained, and blood samples were collected at the third trimester of pregnancy; also, on 45 NBs (20 GDM and 25 controls) from whom umbilical cord blood samples were obtained. Humoral immunity was analyzed by measuring anti-T. gondii total IgG, IgG subclasses and IgG avidity. To evaluate cellular immunity, peripheral blood mononuclear cells (PBMC) from 32 PW (16 GDM and 16 controls) were cultured, supernatant cytokines were determined, and flow cytometry was performed to analyze the expression at lymphocytes of surface molecules, cytokines and transcription factors. All PW and NBs were positive for total IgG, and the prevalent subclass was IgG1. There was a negative correlation between the OGTT glycemia of PW and the levels of total IgG, IgG1 and IgG avidity. The IgG avidity of the GDM group was significantly lower than the control group. Patients from the GDM group had a higher number of T lymphocytes expressing markers of cell activation and exhaustion (CD28 and PD-1). In the presence of T. gondii soluble antigen (STAg) the amount of CD4+ T cells producing IFN-γ, IL-10 and IL-17 was significantly lower in the GDM group, while there was no difference between groups in the number of CD4+ CD25HighFOXP3+LAP+ functional Treg cells. Additionally, under STAg stimulus, the secretion of IL-17, IL-4, TNF and IL-2 cytokines at PBMCs culture supernatant was lower in the GDM group. In conclusion, there was a correlation between the increase in blood glucose and the decrease in levels of anti-T. gondii antibodies, associated with the decreased IgG avidity in patients who develop GDM. Also, the GDM group had decreased immune responses in Th1, Th2 and Th17 profiles, suggesting an association between GDM and the negative modulation of the humoral and cellular immune responses against T. gondii.
Collapse
Affiliation(s)
- Ana Carolina de Morais Oliveira-Scussel
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Paula Tatiana Mutão Ferreira
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Renata de Souza Resende
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Cristhianne Molinero Ratkevicius-Andrade
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Angelica de Oliveira Gomes
- Laboratory of Cellular Interactions, Institute of Biological and Natural Sciences, Department of Structural Biology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marina Carvalho Paschoini
- Institute of Health Sciences, Department of Obstetricy, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Fernanda Bernadelli De Vito
- Laboratory of Hematology and Hemotherapy, Institute of Health Sciences, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Thaís Soares Farnesi-de-Assunção
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Marcos Vinícius da Silva
- Laboratory of Parasitology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - José Roberto Mineo
- Laboratory of Immunology “Dr. Mário Endsfeldz Camargo”, Institute of Biomedical Sciences, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | | | - Virmondes Rodrigues
- Laboratory of Immunology, Institute of Biological and Natural Sciences, Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| |
Collapse
|
18
|
Devvanshi H, Kachhwaha R, Manhswita A, Bhatnagar S, Kshetrapal P. Immunological Changes in Pregnancy and Prospects of Therapeutic Pla-Xosomes in Adverse Pregnancy Outcomes. Front Pharmacol 2022; 13:895254. [PMID: 35517798 PMCID: PMC9065684 DOI: 10.3389/fphar.2022.895254] [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] [Received: 03/13/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Stringent balance of the immune system is a key regulatory factor in defining successful implantation, fetal development, and timely parturition. Interference in these primary regulatory mechanisms, either at adolescence or prenatal state led to adverse pregnancy outcomes. Fertility restoration with the help of injectable gonadotrophins/progesterone, ovulation-inducing drugs, immunomodulatory drugs (corticosteroids), and reproductive surgeries provides inadequate responses, which manifest its own side effects. The development of a potential diagnostic biomarker and an effectual treatment for adverse pregnancy outcomes is a prerequisite to maternal and child health. Parent cell originated bi-layered-intraluminal nano-vesicles (30-150 nm) also known as exosomes are detected in all types of bodily fluids like blood, saliva, breast milk, urine, etc. Exosomes being the most biological residual structures with the least cytotoxicity are loaded with cargo in the form of RNAs (miRNAs), proteins (cytokines), hormones (estrogen, progesterone, etc.), cDNAs, and metabolites making them chief molecules of cell-cell communication. Their keen involvement in the regulation of biological processes has portrayed them as the power shots of cues to understand the disease's pathophysiology and progression. Recent studies have demonstrated the role of immunexosomes (immunomodulating exosomes) in maintaining unwavering immune homeostasis between the mother and developing fetus for a healthy pregnancy. Moreover, the concentration and size of the exosomes are extensively studied in adverse pregnancies like preeclampsia, gestational diabetes mellitus (GDM), and preterm premature rupture of membrane (pPROMs) as an early diagnostic marker, thus giving in-depth information about their pathophysiology. Exosomes have also been engineered physically as well as genetically to enhance their encapsulation efficiency and specificity in therapy for cancer and adverse pregnancies. Successful bench to bedside discoveries and interventions in cancer has motivated developmental biologists to investigate the role of immunexosomes and their active components. Our review summarizes the pre-clinical studies for the use of these power-shots as therapeutic agents. We envisage that these studies will pave the path for the use of immunexosomes in clinical settings for reproductive problems that arise due to immune perturbance in homeostasis either at adolescence or prenatal state.
Collapse
Affiliation(s)
- Himadri Devvanshi
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Rohit Kachhwaha
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Anima Manhswita
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD, Australia
| | - Shinjini Bhatnagar
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
19
|
Dynamic changes in regulatory T cells during normal pregnancy, recurrent pregnancy loss, and gestational diabetes. J Reprod Immunol 2022; 150:103492. [DOI: 10.1016/j.jri.2022.103492] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
20
|
Zhang J, Chen J, Gao C, Sun X, Wang L, Hu Z, Li G, Wang J, Wang A. Maggot treatment promotes healing of diabetic foot ulcer wounds possibly by upregulating Treg levels. Diabetes Res Clin Pract 2022; 184:109187. [PMID: 35016990 DOI: 10.1016/j.diabres.2021.109187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 11/30/2021] [Accepted: 12/23/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE Through the study of regulatory T cells (Tregs), we found a possible way to promote the healing of diabetic foot ulcers (DFUs) with maggot treatment and investigated the associated mechanism. METHODS Immunohistochemistry was used to examinetissues from DFU patients treated with or without maggot debridement therapy (MDT). The expression of the signature Treg molecule Foxp3, interleukin-10 (IL-10), transforming growth factor-beta (TGF-β), and interferon regulatory factor 4 (IRF-4) in patients with DFU treated with or without MDT was tested by real-time PCR (RT-PCR). CD4+ T cells from mouse spleen cells were cocultured in vitro with maggot excretions/secretions (ES), and Foxp3, IL-10, TGF-β, and IRF-4 levels were measured by RT-PCR. RESULTS Foxp3 expression was obviously increased in DFU patients treated using MDT but less pronounced in those treated without MDT (P < 0.05). Foxp3, IL-10, TGF-β, and IRF-4 gene expression levels were higher in DFU patients treated with MDT than in those treated without MDT. Moreover, in vitro coculture of mouse spleen cells with ESs produced results consistent with the in vivo results (P < 0.001). CONCLUSION MDT/ESs can obviously upregulate the Treg level and may affect DFU healing in different ways, suggesting a new direction for the future treatment of DFU.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China; Nanjing Prevention and Treatment Center for Occupational Diseases, Nanjing, Jiangsu Province, China
| | - Jin'an Chen
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Chunchen Gao
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Xinjuan Sun
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Lei Wang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Zhiwei Hu
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Gai Li
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China
| | - Jing Wang
- Translational Medicine Center, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China.
| | - Aiping Wang
- Department of Endocrinology, Nanjing JunXie Hospital, Nanjing, Jiangsu Province, China.
| |
Collapse
|
21
|
Sharma S, Banerjee S, Krueger PM, Blois SM. Immunobiology of Gestational Diabetes Mellitus in Post-Medawar Era. Front Immunol 2022; 12:758267. [PMID: 35046934 PMCID: PMC8761800 DOI: 10.3389/fimmu.2021.758267] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Although the concepts related to fetal immune tolerance proposed by Sir Peter Medawar in the 1950s have not withstood the test of time, they revolutionized our current understanding of the immunity at the maternal-fetal interface. An important extension of the original Medawar paradigm is the investigation into the underlying mechanisms for adverse pregnancy outcomes, including recurrent spontaneous abortion, preterm birth, preeclampsia and gestational diabetes mellitus (GDM). Although a common pregnancy complication with systemic symptoms, GDM still lacks understanding of immunological perturbations associated with the pathological processes, particularly at the maternal-fetal interface. GDM has been characterized by low grade systemic inflammation that exacerbates maternal immune responses. In this regard, GDM may also entail mild autoimmune pathology by dysregulating circulating and uterine regulatory T cells (Tregs). The aim of this review article is to focus on maternal-fetal immunological tolerance phenomenon and discuss how local or systemic inflammation has been programmed in GDM. Specifically, this review addresses the following questions: Does the inflammatory or exhausted Treg population affecting the Th17:Treg ratio lead to the propensity of a pro-inflammatory environment? Do glycans and glycan-binding proteins (mainly galectins) contribute to the biology of immune responses in GDM? Our understanding of these important questions is still elementary as there are no well-defined animal models that mimic all the features of GDM or can be used to better understand the mechanistic underpinnings associated with this common pregnancy complication. In this review, we will leverage our preliminary studies and the literature to provide a conceptualized discussion on the immunobiology of GDM.
Collapse
Affiliation(s)
- Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Sayani Banerjee
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Paula M Krueger
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
22
|
Rola autoimmunizacji w rozwoju powikłań cukrzycowych – przegląd badań. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Przewlekłe powikłania cukrzycy są główną przyczyną obniżenia jakości życia, niepełnosprawności, a nawet przedwczesnej śmierci pacjentów cierpiących na tę chorobę. Mimo istotnego postępu w dziedzinie farmakoterapii, ich leczenie pozostaje nadal wyzwaniem w codziennej praktyce klinicznej. Brak terapii przyczynowej wynika z niewystarczającego zrozumienia molekularnych mechanizmów uszkadzających poszczególne narządy w cukrzycy. Uważa się, że etiopatogeneza tych powikłań jest złożona i zależy od czynników genetycznych i środowiskowych. W ich rozwoju, oprócz zaburzeń metabolicznych związanych z hiperglikemią, nasilenia stresu oksydacyjnego, dysfunkcji śródbłonka, indukcji stanu zapalnego, coraz częściej wskazuje się też na znaczącą rolę zaburzeń immunologicznych.
Wyniki badań doświadczalnych przeprowadzonych na zwierzętach, jak również na hodowlach tkankowych, oraz obserwacje kliniczne potwierdzają udział układu odpornościowego obejmujący aktywność autoreaktywnych limfocytów oraz cytotoksyczne działanie autoprzeciwciał w rozwoju poszczególnych powikłań w obu typach cukrzycy. Wydaje się zatem, że zachwianie równowagi immunologicznej wyzwalające autoagresję jest ważnym czynnikiem przyczyniającym się do dysfunkcji poszczególnych organów w typach cukrzycy 1 i 2.
Dokładne zrozumienie immunopatogenezy tych zaburzeń może zmienić dotychczasowe podejście w leczeniu powikłań cukrzycy oraz umożliwić opracowanie skutecznej terapii przyczynowej ukierunkowanej na układ odpornościowy. Identyfikacja swoistych autoprzeciwciał mogłaby usprawnić ich wczesną diagnostykę i prewencję. W artykule podjęto próbę analizy czynników ryzyka najczęstszych schorzeń o podłożu autoimmunizacyjnym, ich związku z typem 1 i 2 cukrzycy oraz podsumowano potencjalne znaczenie autoagresji w rozwoju jej powikłań w oparciu o wyniki dotychczasowych badań doświadczalnych i klinicznych.
Collapse
|
23
|
Olmos-Ortiz A, Flores-Espinosa P, Díaz L, Velázquez P, Ramírez-Isarraraz C, Zaga-Clavellina V. Immunoendocrine Dysregulation during Gestational Diabetes Mellitus: The Central Role of the Placenta. Int J Mol Sci 2021; 22:8087. [PMID: 34360849 PMCID: PMC8348825 DOI: 10.3390/ijms22158087] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Gestational Diabetes Mellitus (GDM) is a transitory metabolic condition caused by dysregulation triggered by intolerance to carbohydrates, dysfunction of beta-pancreatic and endothelial cells, and insulin resistance during pregnancy. However, this disease includes not only changes related to metabolic distress but also placental immunoendocrine adaptations, resulting in harmful effects to the mother and fetus. In this review, we focus on the placenta as an immuno-endocrine organ that can recognize and respond to the hyperglycemic environment. It synthesizes diverse chemicals that play a role in inflammation, innate defense, endocrine response, oxidative stress, and angiogenesis, all associated with different perinatal outcomes.
Collapse
Affiliation(s)
- Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer), Ciudad de México 11000, Mexico; (A.O.-O.); (P.F.-E.)
| | - Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer), Ciudad de México 11000, Mexico; (A.O.-O.); (P.F.-E.)
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico;
| | - Pilar Velázquez
- Departamento de Ginecología y Obstetricia, Hospital Ángeles México, Ciudad de México 11800, Mexico;
| | - Carlos Ramírez-Isarraraz
- Clínica de Urología Ginecológica, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer), Ciudad de México 11000, Mexico;
| | - Verónica Zaga-Clavellina
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer), Ciudad de México 11000, Mexico
| |
Collapse
|
24
|
Munro SK, Balakrishnan B, Lissaman AC, Gujral P, Ponnampalam AP. Cytokines and pregnancy: Potential regulation by histone deacetylases. Mol Reprod Dev 2021; 88:321-337. [PMID: 33904218 DOI: 10.1002/mrd.23430] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/26/2022]
Abstract
Cytokines are important regulators of pregnancy and parturition. Aberrant expression of proinflammatory cytokines during pregnancy contributes towards preterm labor, pre-eclampsia, and gestational diabetes mellitus. The regulation of cytokine expression in human cells is highly complex, involving interactions between environment, transcription factors, and feedback mechanisms. Recent developments in epigenetic research have made tremendous advancements in exploring histone modifications as a key epigenetic regulator of cytokine expression and the effect of their signaling molecules on various organ systems in the human body. Histone acetylation and subsequent deacetylation by histone deacetylases (HDACs) are major epigenetic regulators of protein expression in the human body. The expression of various proinflammatory cytokines, their role in normal and abnormal pregnancy, and their epigenetic regulation via HDACs will be discussed in this review.
Collapse
Affiliation(s)
- Sheryl K Munro
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Biju Balakrishnan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Abbey C Lissaman
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Palak Gujral
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Anna P Ponnampalam
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Physiology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
McElwain CJ, McCarthy FP, McCarthy CM. Gestational Diabetes Mellitus and Maternal Immune Dysregulation: What We Know So Far. Int J Mol Sci 2021; 22:4261. [PMID: 33923959 PMCID: PMC8073796 DOI: 10.3390/ijms22084261] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is an obstetric complication that affects approximately 5-10% of all pregnancies worldwide. GDM is defined as any degree of glucose intolerance with onset or first recognition during pregnancy, and is characterized by exaggerated insulin resistance, a condition which is already pronounced in healthy pregnancies. Maternal hyperglycaemia ensues, instigating a 'glucose stress' response and concurrent systemic inflammation. Previous findings have proposed that both placental and visceral adipose tissue play a part in instigating and mediating this low-grade inflammatory response which involves altered infiltration, differentiation and activation of maternal innate and adaptive immune cells. The resulting maternal immune dysregulation is responsible for exacerbation of the condition and a further reduction in maternal insulin sensitivity. GDM pathology results in maternal and foetal adverse outcomes such as increased susceptibility to diabetes mellitus development and foetal neurological conditions. A clearer understanding of how these pathways originate and evolve will improve therapeutic targeting. In this review, we will explore the existing findings describing maternal immunological adaption in GDM in an attempt to highlight our current understanding of GDM-mediated immune dysregulation and identify areas where further research is required.
Collapse
Affiliation(s)
- Colm J. McElwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland;
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, T12 YE02 Cork, Ireland;
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland;
| |
Collapse
|
26
|
CD28null and Regulatory T Cells Are Substantially Disrupted in Patients with End-Stage Renal Disease Due to Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22062975. [PMID: 33804135 PMCID: PMC8001943 DOI: 10.3390/ijms22062975] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Background: End-stage renal disease (ESRD) is associated with alterations in T-cell immunity, including increased CD28null and reduced regulatory T cells (Tregs). However, whether immune disturbances are due to ESRD or primary disease is not yet clear. As diabetes mellitus is the leading cause of ESRD, we evaluated its impact on the immune profile of ESRD patients. Methods: CD28null, Tregs, and natural killer cells were initially analyzed by flow cytometry in 30 predialysis ESRD patients due to diabetes (DM), 30 non-DM (NDM), and 25 healthy controls. Measurements were repeated after 6 months on hemodialysis (HD) or peritoneal dialysis (CAPD). Results: The percentage of CD4 + CD28null cells, CD8 + CD28null cells, and Tregs showed significant differences in DM, NDM, and controls; mean rank 33.71 vs. 25.68 vs. 18.88, p = 0.006, 37.79 vs. 28.82 vs. 17.08, p = 0.008, and 20.79 vs. 26.12 vs. 41.33, p = 0.001, respectively. DM vs. NDM had increased CD4 + CD28null and CD8 + CD28null cells, 11.5% (1.5%–24%) vs. 4.1% (0–42.3%), p = 0.02 and 61.3% (24%–76%) vs. 43% (5.7%–85%), p = 0.04, respectively. After 6 months on HD but not CAPD, DM showed a significant further increase in CD4 + CD28null cells, from 30 (14–100) to 52.7 (15–203), p = 0.02; and CD8 + CD28null cells, from 137 (56–275) to 266 (103–456), p = 0.01. Conclusions: Diabetes mellitus affects T-cell subtypes even at predialysis stage, though changes become more prominent after commencement on HD.
Collapse
|
27
|
Heindel JJ, Belcher S, Flaws JA, Prins GS, Ho SM, Mao J, Patisaul HB, Ricke W, Rosenfeld CS, Soto AM, Vom Saal FS, Zoeller RT. Data integration, analysis, and interpretation of eight academic CLARITY-BPA studies. Reprod Toxicol 2020; 98:29-60. [PMID: 32682780 PMCID: PMC7365109 DOI: 10.1016/j.reprotox.2020.05.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/03/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
"Consortium Linking Academic and Regulatory Insights on BPA Toxicity" (CLARITY-BPA) was a comprehensive "industry-standard" Good Laboratory Practice (GLP)-compliant 2-year chronic exposure study of bisphenol A (BPA) toxicity that was supplemented by hypothesis-driven independent investigator-initiated studies. The investigator-initiated studies were focused on integrating disease-associated, molecular, and physiological endpoints previously found by academic scientists into an industry standard guideline-compliant toxicity study. Thus, the goal of this collaboration was to provide a more comprehensive dataset upon which to base safety standards and to determine whether industry-standard tests are as sensitive and predictive as molecular and disease-associated endpoints. The goal of this report is to integrate the findings from the investigator-initiated studies into a comprehensive overview of the observed impacts of BPA across the multiple organs and systems analyzed. For each organ system, we provide the rationale for the study, an overview of methodology, and summarize major findings. We then compare the results of the CLARITY-BPA studies across organ systems with the results of previous peer-reviewed studies from independent labs. Finally, we discuss potential influences that contributed to differences between studies. Developmental exposure to BPA can lead to adverse effects in multiple organs systems, including the brain, prostate gland, urinary tract, ovary, mammary gland, and heart. As published previously, many effects were at the lowest dose tested, 2.5μg/kg /day, and many of the responses were non-monotonic. Because the low dose of BPA affected endpoints in the same animals across organs evaluated in different labs, we conclude that these are biologically - and toxicologically - relevant.
Collapse
Affiliation(s)
- Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies Commonweal, Bolinas, CA 94924, United States.
| | - Scott Belcher
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Gail S Prins
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago IL 60612, United States
| | - Shuk-Mei Ho
- Department of Environmental Health, University of Cincinnati, Cincinnati OH 45267, United States; Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Jiude Mao
- Biomedical Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - William Ricke
- Department of Urology, University of Wisconsin, Madison WI 53705, United States
| | - Cheryl S Rosenfeld
- Biomedical Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Ana M Soto
- Tufts University, Boston, MA 02111, United States
| | - Frederick S Vom Saal
- Department of Biology, University of Missouri, Columbia, MO 65211, United States
| | - R Thomas Zoeller
- Department of Biology, University of Massachusetts, Amherst, MA 01003, United States
| |
Collapse
|
28
|
St-Germain LE, Castellana B, Baltayeva J, Beristain AG. Maternal Obesity and the Uterine Immune Cell Landscape: The Shaping Role of Inflammation. Int J Mol Sci 2020; 21:E3776. [PMID: 32471078 PMCID: PMC7312391 DOI: 10.3390/ijms21113776] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is often equated to the physiological response to injury or infection. Inflammatory responses defined by cytokine storms control cellular mechanisms that can either resolve quickly (i.e., acute inflammation) or remain prolonged and unabated (i.e., chronic inflammation). Perhaps less well-appreciated is the importance of inflammatory processes central to healthy pregnancy, including implantation, early stages of placentation, and parturition. Pregnancy juxtaposed with disease can lead to the perpetuation of aberrant inflammation that likely contributes to or potentiates maternal morbidity and poor fetal outcome. Maternal obesity, a prevalent condition within women of reproductive age, associates with increased risk of developing multiple pregnancy disorders. Importantly, chronic low-grade inflammation is thought to underlie the development of obesity-related obstetric and perinatal complications. While diverse subsets of uterine immune cells play central roles in initiating and maintaining healthy pregnancy, uterine leukocyte dysfunction as a result of maternal obesity may underpin the development of pregnancy disorders. In this review we discuss the current knowledge related to the impact of maternal obesity and obesity-associated inflammation on uterine immune cell function, utero-placental establishment, and pregnancy health.
Collapse
Affiliation(s)
- Lauren E. St-Germain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Barbara Castellana
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Jennet Baltayeva
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| | - Alexander G. Beristain
- The British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; (L.E.S.-G.); (B.C.); (J.B.)
- Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC V6Z 2K8, Canada
| |
Collapse
|
29
|
De Luccia TPB, Pendeloski KPT, Ono E, Mattar R, Pares DBS, Yazaki Sun S, Daher S. Unveiling the pathophysiology of gestational diabetes: Studies on local and peripheral immune cells. Scand J Immunol 2020; 91:e12860. [PMID: 31849072 DOI: 10.1111/sji.12860] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/10/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022]
Abstract
Gestational diabetes mellitus (GDM) has been associated with impaired maternal immune response. Our aim was to review the available literature linking immune cells profile to GDM, in order to comprehend the role that different subpopulations play in the development of this pathology. We searched in PubMed for studies published in the last decade on circulating levels and placenta expression of immune cells on GDM. We identified 18 studies with several differences regarding the study design, clinical characteristics, number of participants, cell subpopulation and type of sample. Most studies assessed only one subpopulation either in peripheral blood or placenta and did not analyse functional properties of the cells. The most frequently evaluated immune cells were T lymphocytes, especially regulatory T (Tregs), and natural killer (NK) cells in the peripheral blood, and placental macrophages. No studies analysing B cells were identified, and only one study each evaluating γδT cells, dendritic cell (DC) and monocytes was found. Although there are controversies, at least one study reported positive association between GDM and CD4+ (activated), Tregs, Th17 and γδT cells; neutrophil/lymphocyte; NK cell (cytotoxic); macrophages; and monocytes. The number of studies is still small, so caution should be exercised in interpreting the data, and further research is required to validate these findings and establish the role of adaptive and innate immune cells in GDM pathophysiology.
Collapse
Affiliation(s)
- Thiago P B De Luccia
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - Karen P T Pendeloski
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - Erika Ono
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - Rosiane Mattar
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - David B S Pares
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - Sue Yazaki Sun
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| | - Silvia Daher
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
30
|
Le Garf S, Murdaca J, Mothe-Satney I, Sibille B, Le Menn G, Chinetti G, Neels JG, Rousseau AS. Complementary Immunometabolic Effects of Exercise and PPARβ/δ Agonist in the Context of Diet-Induced Weight Loss in Obese Female Mice. Int J Mol Sci 2019; 20:E5182. [PMID: 31635041 PMCID: PMC6829333 DOI: 10.3390/ijms20205182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
Regular aerobic exercise, independently of weight loss, improves metabolic and anti-inflammatory states, and can be regarded as beneficial in counteracting obesity-induced low-grade inflammation. However, it is still unknown how exercise alters immunometabolism in a context of dietary changes. Agonists of the Peroxisome Proliferator Activated-Receptor beta/delta (PPARβ/δ) have been studied this last decade as "exercise-mimetics", which are potential therapies for metabolic diseases. In this study, we address the question of whether PPARβ/δ agonist treatment would improve the immunometabolic changes induced by exercise in diet-induced obese female mice, having switched from a high fat diet to a normal diet. 24 mice were assigned to groups according to an 8-week exercise training program and/or an 8-week treatment with 3 mg/kg/day of GW0742, a PPARβ/δ agonist. Our results show metabolic changes of peripheral lymphoid tissues with PPARβ/δ agonist (increase in fatty acid oxidation gene expression) or exercise (increase in AMPK activity) and a potentiating effect of the combination of both on the percentage of anti-inflammatory Foxp3+ T cells. Those effects are associated with a decreased visceral adipose tissue mass and skeletal muscle inflammation (TNF-α, Il-6, Il-1β mRNA level), an increase in skeletal muscle oxidative capacities (citrate synthase activity, endurance capacity), and insulin sensitivity. We conclude that a therapeutic approach targeting the PPARβ/δ pathway would improve obesity treatment.
Collapse
Affiliation(s)
| | - Joseph Murdaca
- Université Côte d'Azur, INSERM, C3M, CEDEX 3, 06204 Nice, France.
| | | | - Brigitte Sibille
- Université Côte d'Azur, INSERM, C3M, CEDEX 3, 06204 Nice, France.
| | | | - Giulia Chinetti
- Université Côte d'Azur, INSERM, C3M, CHU, CEDEX 3, 06204 Nice, France.
| | - Jaap G Neels
- Université Côte d'Azur, INSERM, C3M, CEDEX 3, 06204 Nice, France.
| | | |
Collapse
|
31
|
Fuller EA, Younesi S, Xavier S, Sominsky L. Neuroimmune regulation of female reproduction in health and disease. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2019.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
32
|
Al-Ofi EA. Implications of inflammation and insulin resistance in obese pregnant women with gestational diabetes: A case study. SAGE Open Med Case Rep 2019; 7:2050313X19843737. [PMID: 31041103 PMCID: PMC6477763 DOI: 10.1177/2050313x19843737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 03/18/2019] [Indexed: 12/21/2022] Open
Abstract
Background Obesity is one of the leading pregnancy risks for both the mother and the neonate. The prevalence of gestational diabetes mellitus has been increasing, especially with the increase in obesity in reproductive-aged women. A high body mass index, a sedentary lifestyle, a previous macrosomic infant, polycystic ovary syndrome and hypothyroidism are the main risk factors for gestational diabetes mellitus. Early gestational diabetes mellitus detection in high-risk individuals is a useful method for preventing further complications and/or preventing this disease by improving the patient's lifestyle. Case presentation A morbidly obese woman with a high body mass index (>36) at 24 weeks gestational age presented with several gestational diabetes mellitus risk factors. Her glucose tolerance test verified gestational diabetes mellitus, and, incidentally, her C-reactive protein level was elevated without obvious reason. Her plasma levels of inflammatory cytokines had also been assessed and were exaggerated. After lifestyle intervention, including weight management, the patient's inflammatory mediators, including her C-reactive protein level, dropped. Therefore, this study aimed to identify the relationship between the patient's inflammation and obesity. Conclusion Antenatal C-reactive protein screening could be used throughout pregnancy to predict inflammation from high-risk pregnant women. This case scenario describes the interrelationships between inflammation, insulin resistance and adipokines, as well as the contributions of hypothyroidism and polycystic ovary syndrome. Further research should emphasise the relationships between inflammation and obesity in pregnancy.
Collapse
Affiliation(s)
- Ebtisam Aziz Al-Ofi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Sallam NA, Palmgren VAC, Singh RD, John CM, Thompson JA. Programming of Vascular Dysfunction in the Intrauterine Milieu of Diabetic Pregnancies. Int J Mol Sci 2018; 19:E3665. [PMID: 30463313 PMCID: PMC6275067 DOI: 10.3390/ijms19113665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023] Open
Abstract
With the rising global tide of obesity, gestational diabetes mellitus (GDM) burgeoned into one of the most common antenatal disorders worldwide. Macrosomic babies born to diabetic mothers are more likely to develop risk factors for cardiovascular disease (CVD) before they reach adulthood. Rodent studies in offspring born to hyperglycemic pregnancies show vascular dysfunction characterized by impaired nitric oxide (NO)-mediated vasodilation and increased production of contractile prostanoids by cyclooxygenase 2 (COX-2). Vascular dysfunction is a key pathogenic event in the progression of diabetes-related vascular disease, primarily attributable to glucotoxicity. Therefore, glucose-induced vascular injury may stem directly from the hyperglycemic intrauterine environment of GDM pregnancy, as evinced by studies showing endothelial activation and inflammation at birth or in childhood in offspring born to GDM mothers. This review discusses potential mechanisms by which intrauterine hyperglycemia programs dysfunction in the developing vasculature.
Collapse
Affiliation(s)
- Nada A Sallam
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Victoria A C Palmgren
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
| | - Radha D Singh
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Cini M John
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
| | - Jennifer A Thompson
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Calgary, AB T2N 4N1, Canada.
- Children's Hospital Research Institute; University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
34
|
Zhou T, Hu Z, Yang S, Sun L, Yu Z, Wang G. Role of Adaptive and Innate Immunity in Type 2 Diabetes Mellitus. J Diabetes Res 2018; 2018:7457269. [PMID: 30533447 PMCID: PMC6250017 DOI: 10.1155/2018/7457269] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022] Open
Abstract
After the recognition of the essential role of the immune system in the progression of type 2 diabetes mellitus, more studies are focused on the effects produced by the abnormal differentiation of components of the immune system. In patients suffering from obesity or T2DM, there were alterations in proliferation of T cells and macrophages, and impairment in function of NK cells and B cells, which represented abnormal innate and adaptive immunity. The abnormality of either innate immunity, adaptive immunity, or both was involved and interacted with each other during the progression of T2DM. Although previous studies have revealed the functional involvement of T cells in T2DM, and the regulation of metabolism by the innate or adaptive immune system during the pathogenesis of T2DM, there has been a lack of literature reviewing the relevant role of adaptive and innate immunity in the progression of T2DM. Here, we will review their relevant roles, aiming to provide new thought for the development of immunotherapy in T2DM.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zheng Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Shuo Yang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhenxiang Yu
- Department of Respiration, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
35
|
The diabetes pandemic and associated infections: suggestions for clinical microbiology. ACTA ACUST UNITED AC 2018; 30:1-17. [PMID: 30662163 PMCID: PMC6319590 DOI: 10.1097/mrm.0000000000000155] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/08/2017] [Indexed: 12/15/2022]
Abstract
There are 425 million people with diabetes mellitus in the world. By 2045, this figure will grow to over 600 million. Diabetes mellitus is classified among noncommunicable diseases. Evidence points to a key role of microbes in diabetes mellitus, both as infectious agents associated with the diabetic status and as possible causative factors of diabetes mellitus. This review takes into account the different forms of diabetes mellitus, the genetic determinants that predispose to type 1 and type 2 diabetes mellitus (especially those with possible immunologic impact), the immune dysfunctions that have been documented in diabetes mellitus. Common infections occurring more frequently in diabetic vs. nondiabetic individuals are reviewed. Infectious agents that are suspected of playing an etiologic/triggering role in diabetes mellitus are presented, with emphasis on enteroviruses, the hygiene hypothesis, and the environment. Among biological agents possibly linked to diabetes mellitus, the gut microbiome, hepatitis C virus, and prion-like protein aggregates are discussed. Finally, preventive vaccines recommended in the management of diabetic patients are considered, including the bacillus calmette-Guerin vaccine that is being tested for type 1 diabetes mellitus. Evidence supports the notion that attenuation of immune defenses (both congenital and secondary to metabolic disturbances as well as to microangiopathy and neuropathy) makes diabetic people more prone to certain infections. Attentive microbiologic monitoring of diabetic patients is thus recommendable. As genetic predisposition cannot be changed, research needs to identify the biological agents that may have an etiologic role in diabetes mellitus, and to envisage curative and preventive ways to limit the diabetes pandemic.
Collapse
|
36
|
Lobo TF, Borges CDM, Mattar R, Gomes CP, de Ângelo AGS, Pendeloski KPT, Daher S. In response to Manuscript AJRI 01-18-016 entitled "Regulatory T cells, natural killer cells and obesity in patients with gestational diabetes mellitus"-Kawada T. Am J Reprod Immunol 2018; 79:e12830. [PMID: 29473718 DOI: 10.1111/aji.12830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
| | | | - Rosiane Mattar
- Department of Obstetrics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caio Perez Gomes
- Department of Obstetrics, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Silvia Daher
- Department of Obstetrics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Kawada T. Regulatory T cells, natural killer cells, and obesity in patients with gestational diabetes mellitus. Am J Reprod Immunol 2018; 79:e12831. [PMID: 29453819 DOI: 10.1111/aji.12831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Tomoyuki Kawada
- Department of Hygiene and Public Health, Nippon Medical School, Tokyo, Japan
| |
Collapse
|